1
|
Liu J, Hanson A, Yin W, Wu Q, Wauthier E, Diao J, Dinh T, Macdonald J, Li R, Terajima M, Yamauchi M, Chen Z, Sethupathy P, Dong J, Reid LM, Wang Y. Decellularized liver scaffolds for constructing drug-metabolically functional ex vivo human liver models. Bioact Mater 2025; 43:162-180. [PMID: 39386220 PMCID: PMC11462156 DOI: 10.1016/j.bioactmat.2024.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
The creation of ex vivo human liver models has long been a critical objective in academic, clinical, and pharmaceutical research, particularly for drug development, where accurate evaluation of hepatic metabolic dynamics is crucial. We have developed a bioengineered, perfused, organ-level human liver model that accurately replicates key liver functions, including metabolic activities, and protein synthesis, thus addressing some of the limitations associated with traditional liver monolayers, organoids, and matrix-embedded liver cells. Our approach utilizes liver-specific biomatrix scaffolds, prepared using an innovative protocol and fortified with matrix components that facilitate cellular interactions. These scaffolds, when seeded with human fetal liver cells or co-seeded with liver parenchymal and endothelial cell lines, enable the formation of three-dimensional (3D) human livers with enhanced cellular organization. The "recellularized tissue-engineered livers" (RCLs) have undergone various analyses, demonstrating the capability for establishing liver microenvironments ex vivo. Within 7-14 days, the RCLs exhibit evidence of liver differentiation and metabolic capabilities, underscoring the potential for use in drug metabolism and toxicity studies. Although our study represents a significant step forward, we acknowledge the need for direct comparisons with existing models and further research to fully elucidate the spectrum of regenerative responses. The high drug-metabolizing enzyme activity of RCLs, as demonstrated in our study, provides a promising avenue for investigating drug-induced liver injury mechanisms, contributing to a more detailed understanding of early drug discovery processes.
Collapse
Affiliation(s)
- Juan Liu
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| | - Ariel Hanson
- Departments of Biomedical Engineering, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Wenzhen Yin
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Qiao Wu
- Infection Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Eliane Wauthier
- Departments of Cell Biology and Physiology, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Jinmei Diao
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| | - Timothy Dinh
- Departments of Genetics, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Jeff Macdonald
- Departments of Biomedical Engineering, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Ruihong Li
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Masahiko Terajima
- Oral and Craniofacial Health Sciences, UNC School of Dentistry, Chapel Hill, NC, 27599, USA
| | - Mitsuo Yamauchi
- Oral and Craniofacial Health Sciences, UNC School of Dentistry, Chapel Hill, NC, 27599, USA
| | - Ziye Chen
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Praveen Sethupathy
- Departments of Genetics, UNC School of Medicine, Chapel Hill, NC, 27599, USA
- Division of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Jiahong Dong
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| | - Lola M. Reid
- Departments of Biomedical Engineering, UNC School of Medicine, Chapel Hill, NC, 27599, USA
- Program in Molecular Biology and Biotechnology, UNC School of Medicine, Chapel Hill, NC, 27599, USA
| | - Yunfang Wang
- Hepato-pancreato-biliary Center, Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
2
|
Bleckwehl T, Babler A, Tebens M, Maryam S, Nyberg M, Bosteen M, Halder M, Shaw I, Fleig S, Pyke C, Hvid H, Voetmann LM, van Buul JD, Sluimer JC, Das V, Baumgart S, Kramann R, Hayat S. Encompassing view of spatial and single-cell RNA sequencing renews the role of the microvasculature in human atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024:10.1038/s44161-024-00582-1. [PMID: 39715784 DOI: 10.1038/s44161-024-00582-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 11/04/2024] [Indexed: 12/25/2024]
Abstract
Atherosclerosis is a pervasive contributor to ischemic heart disease and stroke. Despite the advance of lipid-lowering therapies and anti-hypertensive agents, the residual risk of an atherosclerotic event remains high, and developing therapeutic strategies has proven challenging. This is due to the complexity of atherosclerosis with a spatial interplay of multiple cell types within the vascular wall. In this study, we generated an integrative high-resolution map of human atherosclerotic plaques combining single-cell RNA sequencing from multiple studies and spatial transcriptomics data from 12 human specimens with different stages of atherosclerosis. Here we show cell-type-specific and atherosclerosis-specific expression changes and spatially constrained alterations in cell-cell communication. We highlight the possible recruitment of lymphocytes via ACKR1 endothelial cells of the vasa vasorum, the migration of vascular smooth muscle cells toward the lumen by transforming into fibromyocytes and cell-cell communication in the plaque region, indicating an intricate cellular interplay within the adventitia and the subendothelial space in human atherosclerosis.
Collapse
Affiliation(s)
- Tore Bleckwehl
- Department of Medicine 2, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Anne Babler
- Department of Medicine 2, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Merel Tebens
- Department of Medical Biochemistry, Vascular Cell Biology Lab at Amsterdam UMC, location AMC, Sanquin Research and Landsteiner Laboratory and Leeuwenhoek Centre for Advanced Microscopy at Swammerdam Institute for Life Sciences at the University of Amsterdam, Amsterdam, The Netherlands
| | - Sidrah Maryam
- Department of Medicine 2, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | | | | | - Maurice Halder
- Department of Medicine 2, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Isaac Shaw
- Department of Medicine 2, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Susanne Fleig
- Department of Medicine 2, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Charles Pyke
- Pathology & Imaging, Global Drug Development, Novo Nordisk A/S, Måløv, Denmark
| | - Henning Hvid
- Pathology & Imaging, Global Drug Development, Novo Nordisk A/S, Måløv, Denmark
| | | | - Jaap D van Buul
- Department of Medical Biochemistry, Vascular Cell Biology Lab at Amsterdam UMC, location AMC, Sanquin Research and Landsteiner Laboratory and Leeuwenhoek Centre for Advanced Microscopy at Swammerdam Institute for Life Sciences at the University of Amsterdam, Amsterdam, The Netherlands
| | - Judith C Sluimer
- Department of Pathology, ARIM School for Cardiovascular Sciences, Maastricht University Medical Center (MUMC), Maastricht, The Netherlands
- BHF Centre for Cardiovascular Sciences (CVS), University of Edinburgh, Edinburgh, UK
| | - Vivek Das
- Digital Science and Innovation, Computational Biology - AI & Digital Research, Novo Nordisk A/S, Måløv, Denmark
| | - Simon Baumgart
- Digital Science and Innovation, Computational Biology - AI & Digital Research, Novo Nordisk A/S, Måløv, Denmark
| | - Rafael Kramann
- Department of Medicine 2, RWTH Aachen University, Medical Faculty, Aachen, Germany.
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Sikander Hayat
- Department of Medicine 2, RWTH Aachen University, Medical Faculty, Aachen, Germany.
| |
Collapse
|
3
|
Anakha J, Prasad YR, Pande AH. Endostatin in disease modulation: From cancer to beyond. Vascul Pharmacol 2024; 158:107459. [PMID: 39708990 DOI: 10.1016/j.vph.2024.107459] [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: 10/09/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Angiogenesis plays a pivotal role in various pathological conditions, making it a key target in therapeutic development. Anti-angiogenic therapies are gaining traction for their potential in treating a range of angiogenesis-dependent diseases. Among these, endogenous angiogenesis inhibitors, particularly endostatin, have garnered significant attention for their therapeutic potential. While extensively studied for its anti-angiogenic effects in cancer, endostatin also exhibits anti-atherosclerotic and anti-fibrotic properties, broadening its therapeutic scope. Despite the successful clinical use of recombinant human endostatin in China for nearly two decades, its broader therapeutic potential remains underexplored. Thus, this review delves into the multifaceted applications of endostatin, examining its role in ocular diseases, inflammation, reproductive disorders, and tumor angiogenesis. Furthermore, it provides a comprehensive overview of its emerging roles beyond angiogenesis, particularly in the context of atherosclerosis and fibroproliferative conditions.
Collapse
Affiliation(s)
- J Anakha
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India.
| | - Yenisetti Rajendra Prasad
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India.
| |
Collapse
|
4
|
Dabravolski SA, Churov AV, Ravani AL, Karimova AE, Luchinkin IG, Sukhorukov VN, Orekhov AN. The role of Epsins in atherosclerosis: From molecular mechanisms to therapeutic applications. Vascul Pharmacol 2024; 158:107457. [PMID: 39672315 DOI: 10.1016/j.vph.2024.107457] [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: 10/07/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
Atherosclerosis is a multifaceted disease characterised by chronic inflammation and vascular remodelling, leading to plaque formation and cardiovascular complications. Recent evidence highlights the critical role of epsins, a family of endocytic proteins, in the pathogenesis of atherosclerosis. This manuscript explores the multifarious functions of epsins in atherosclerosis, focusing on their involvement in angiogenesis, lymphangiogenesis, and the modulation of key signalling pathways. We discuss how epsins facilitate EndoMT through their interaction with the TGFβ signalling pathway, which contributes to vascular smooth muscle cell-like phenotypes and plaque instability. Additionally, we examine the therapeutic potential of targeting epsins, elucidating their interactions with crucial partners such as LDLR, LRP-1, and TLR 2/4, among others, in mediating lipid metabolism and inflammation. Furthermore, we highlight the promising prospects of epsin-targeting peptides and small interfering RNAs as therapeutic agents for atherosclerosis treatment. Despite these advancements, the research faces limitations, including a reliance on specific mouse models and a need for comprehensive studies on the long-term effects of epsin modulation. Therefore, future investigations should focus on elucidating the detailed mechanisms of epsin function and their implications in cardiovascular health, fostering collaborations to translate basic research into innovative therapeutic strategies. This work underscores the necessity for further exploration of epsins to unlock their full therapeutic potential in combating atherosclerosis and related cardiovascular diseases.
Collapse
Affiliation(s)
- Siarhei A Dabravolski
- Department of Biotechnology Engineering, Braude Academic College of Engineering, Snunit 51, P.O. Box 78, Karmiel 2161002, Israel.
| | - Alexey V Churov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, Russia; Pirogov Russian National Research Medical University, Russia Gerontology Clinical Research Centre, Moscow, Institute on Ageing Research, Russian Federation, 16 1st Leonova Street, 129226 Moscow, Russia
| | - Alessio L Ravani
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, 121609 Moscow, Russia
| | - Amina E Karimova
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, 33, Profsoyuznaya Street, Building 4, 117418 Moscow, Russia
| | - Igor G Luchinkin
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, Russia
| | - Vasily N Sukhorukov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, Russia; Institute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, 119991 Moscow, Russia
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, Russia
| |
Collapse
|
5
|
Xie BL, Song BC, Liu MW, Wen W, Yan YX, Gao MJ, Jiang LL, Jin ZD, Yang L, Liu JG, Shi DZ, Zhao FH. Zedoarondiol Inhibits Neovascularization in Atherosclerotic Plaques of ApoE -/- Mice by Reducing Platelet Exosomes-Derived MiR-let-7a. Chin J Integr Med 2024:10.1007/s11655-024-4003-2. [PMID: 39641887 DOI: 10.1007/s11655-024-4003-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE To investigate the effect of zedoarondiol on neovascularization of atherosclerotic (AS) plaque by exosomes experiment. METHODS ApoE-/- mice were fed with high-fat diet to establish AS model and treated with high- and low-dose (10, 5 mg/kg daily) of zedoarondiol, respectively. After 14 weeks, the expressions of anti-angiogenic protein thrombospondin 1 (THBS-1) and its receptor CD36 in plaques, as well as platelet activation rate and exosome-derived miR-let-7a were detected. Then, zedoarondiol was used to intervene in platelets in vitro, and miR-let-7a was detected in platelet-derived exosomes (Pexo). Finally, human umbilical vein endothelial cells (HUVECs) were transfected with miR-let-7a mimics and treated with Pexo to observe the effect of miR-let-7a in Pexo on tube formation. RESULTS Animal experiments showed that after treating with zedoarondiol, the neovascularization density in plaques of AS mice was significantly reduced, THBS-1 and CD36 increased, the platelet activation rate was markedly reduced, and the miR-let-7a level in Pexo was reduced (P<0.01). In vitro experiments, the platelet activation rate and miR-let-7a levels in Pexo were significantly reduced after zedoarondiol's intervention. Cell experiments showed that after Pexo's intervention, the tube length increased, and the transfection of miR-let-7a minics further increased the tube length of cells, while reducing the expressions of THBS-1 and CD36. CONCLUSION Zedoarondiol has the effect of inhibiting neovascularization within plaque in AS mice, and its mechanism may be potentially related to inhibiting platelet activation and reducing the Pexo-derived miRNA-let-7a level.
Collapse
Affiliation(s)
- Bei-Li Xie
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Bo-Ce Song
- Cardiovascular Department, Beijing Hospital of Integrated Chinese and Western Medicine, Beijing, 100091, China
| | - Ming-Wang Liu
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Wei Wen
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yu-Xin Yan
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Meng-Jie Gao
- Graduate School of Beijing University of Chinese Medicine, Beijing, 100091, China
| | - Lu-Lian Jiang
- Graduate School of Beijing University of Chinese Medicine, Beijing, 100091, China
| | - Zhi-Die Jin
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Lin Yang
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Jian-Gang Liu
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Da-Zhuo Shi
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Fu-Hai Zhao
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China.
| |
Collapse
|
6
|
Zhao Y, Zheng G, Yang S, Liu S, Wu Y, Miao Y, Liang Z, Hua Y, Zhang J, Shi J, Li D, Cheng Y, Zhang Y, Chen Y, Fan G, Ma C. The plant extract PNS mitigates atherosclerosis via promoting Nrf2-mediated inhibition of ferroptosis through reducing USP2-mediated Keap1 deubiquitination. Br J Pharmacol 2024; 181:4822-4844. [PMID: 39228119 DOI: 10.1111/bph.17311] [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: 12/14/2023] [Revised: 06/04/2024] [Accepted: 06/23/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND AND PURPOSE Atherosclerosis is the basis of cardiovascular disease. Ferroptosis is a form of programmed cell death characterized by lipid peroxidation, which contributes to atherogenesis. The plant extract PNS (Panax notoginseng saponins), containing the main active ingredients of Panax notoginseng, exhibits anti-atherogenic properties. Herein, we determined whether PNS and its major components could attenuate atherosclerosis by suppressing ferroptosis and revealed the underlying mechanism(s). EXPERIMENTAL APPROACH The anti-atherogenic effects of PNS and their association with inhibition of ferroptosis was determined in apoE-/- mice. In vitro, the anti-ferroptotic effect and mechanism(s) of PNS components were demonstrated in the presence of ferroptosis inducers. Expression of ferroptosis markers and the ubiquitination of Keap1 were evaluated in USP2-/- macrophages. Finally, the anti-atherogenic effect of USP2 knockout was determined by using USP2-/- mice treated with high-fat diet (HFD) and AAV-PCSK9. KEY RESULTS PNS inhibited ferroptosis and atherosclerosis in vivo. PNS suppressed ferroptosis and ferroptosis-aggravated foam cell formation and inflammation in vitro. Mechanistically, PNS and its components activated Nrf2 by antagonizing Keap1, which was attributed to the inhibition of USP2 expression. USP2 knockout antagonized ferroptosis and ferroptosis-aggravated foam cell formation and inflammation, thus mitigating atherosclerosis. USP2 knockout abolished inhibitory effects of PNS on foam cell formation and inflammation in vitro. CONCLUSION AND IMPLICATIONS PNS reduced USP2-mediated Keap1 de-ubiquitination and promoted Keap1 degradation, thereby activating Nrf2, improving iron metabolism and reducing lipid peroxidation, thus contributing to an anti-atherosclerotic outcome. Our study revealed the mechanism(s) underlying inhibition of ferroptosis and atherosclerosis by PNS.
Collapse
Affiliation(s)
- Yun Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Guobin Zheng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Shu Yang
- Department of Geriatrics, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Shangjing Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yifan Wu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yaodong Miao
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhen Liang
- Department of Geriatrics, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jing Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jia Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Dan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yanfei Cheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yunsha Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yuanli Chen
- Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
7
|
Ajoolabady A, Pratico D, Lin L, Mantzoros CS, Bahijri S, Tuomilehto J, Ren J. Inflammation in atherosclerosis: pathophysiology and mechanisms. Cell Death Dis 2024; 15:817. [PMID: 39528464 PMCID: PMC11555284 DOI: 10.1038/s41419-024-07166-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/26/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Atherosclerosis imposes a heavy burden on cardiovascular health due to its indispensable role in the pathogenesis of cardiovascular disease (CVD) such as coronary artery disease and heart failure. Ample clinical and experimental evidence has corroborated the vital role of inflammation in the pathophysiology of atherosclerosis. Hence, the demand for preclinical research into atherosclerotic inflammation is on the horizon. Indeed, the acquisition of an in-depth knowledge of the molecular and cellular mechanisms of inflammation in atherosclerosis should allow us to identify novel therapeutic targets with translational merits. In this review, we aimed to critically discuss and speculate on the recently identified molecular and cellular mechanisms of inflammation in atherosclerosis. Moreover, we delineated various signaling cascades and proinflammatory responses in macrophages and other leukocytes that promote plaque inflammation and atherosclerosis. In the end, we highlighted potential therapeutic targets, the pros and cons of current interventions, as well as anti-inflammatory and atheroprotective mechanisms.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Ling Lin
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | | | - Suhad Bahijri
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Jaakko Tuomilehto
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia.
- Department of Public Health, University of Helsinki, Helsinki, Finland.
- Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, Finland.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
8
|
Ahmed B, Farb MG, Gokce N. Cardiometabolic implications of adipose tissue aging. Obes Rev 2024; 25:e13806. [PMID: 39076025 DOI: 10.1111/obr.13806] [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: 08/21/2023] [Revised: 03/14/2024] [Accepted: 07/05/2024] [Indexed: 07/31/2024]
Abstract
Adipose tissue is a large endocrine organ that serves numerous physiological functions. As we age, adipose tissue remodels and can develop functional changes that alters its phenotype, potentially contributing to metabolic and cardiovascular disorders. Aging adipose tissue is characterized by regional redistribution of fat, accumulation of senescent cells, fibrosis, and decline in adipocyte differentiation capacities, which collectively impact adipose tissue function and whole body health. A notable transformation involves increased accumulation of intra-abdominal visceral adipose tissue and ectopic fat around internal organs such as the heart, blood vessels, liver, and kidneys that alter their functions. Other changes associated with aging include alterations in adipokine secretion and changes in adipocyte size and numbers. Aging adipocytes play a role in mediating chronic inflammation, metabolic dysfunction, and insulin resistance. Visceral adipose tissue, which increases in volume with aging, is in particular associated with inflammation, angiogenic dysfunction, and microvascular abnormalities, and mediators released by visceral fat may have adverse consequences systemically in multiple target organs, including the cardiovascular system. Understanding mechanisms underlying adipose tissue aging and its impact on cardiovascular health are important for developing interventions and treatments to promote healthy aging and reduce cardiometabolic disease risk.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Melissa G Farb
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Noyan Gokce
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Ryu JY, Chang YJ, Lee JS, Choi KY, Yang JD, Lee SJ, Lee J, Huh S, Kim JY, Chung HY. Extracranial Vascular Malformations Increase Cardiovascular Disease Risk: A Nationwide Population-Based Cohort Study. Plast Reconstr Surg 2024; 154:1047e-1058e. [PMID: 38232222 PMCID: PMC11512613 DOI: 10.1097/prs.0000000000011297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
BACKGROUND Extracranial vascular malformations affect vessel inflammation, clotting, and ischemia. However, the relationship between extracranial vascular malformations and myocardial infarction (MI) or stroke has not been fully elucidated. Limited studies have investigated the association between extracranial vascular malformations and cardiovascular diseases. METHODS A total of 48,701 patients with extracranial vascular malformations and a control cohort of 487,010 age- and sex-matched participants from the Korean National Health Insurance database were included. The incidence and risk of MI, ischemic stroke (IS), and hemorrhagic stroke (HS) between participants with extracranial vascular malformations and the control cohort was compared. RESULTS After adjusting for other cardiovascular disease risk factors, the adjusted hazard ratios (aHRs) for venous malformations, capillary malformations (CMs), arteriovenous malformations (AVMs), and lymphatic malformations in patients with acute MI were 1.25 (CI, 1.04 to 1.50), 1.41 (CI, 1.24 to 1.61), 1.68 (CI, 1.18 to 2.37), and 1.40 (CI, 1.31 to 1.48), respectively. For IS, the aHRs were 1.55 (CI, 1.35 to 1.77), 1.92 (CI, 1.74 to 2.11), 1.13 (CI, 0.78 to 1.64), and 1.51 (CI, 1.44 to 1.58), respectively. For HS, the aHRs were 1.51 (CI, 1.12 to 2.05), 5.63 (CI, 4.97 to 6.38), 2.93 (CI, 1.82 to 4.72), and 1.34 (CI, 1.20 to 1.50), respectively. CONCLUSIONS Independent of cardiovascular risk factors, extracranial vascular malformations were associated with an increased risk of MI, IS, and HS. For patients with CMs and AVMs, intracerebral hemorrhage risk was particularly high, accounting for 563% and 293%, respectively. Therefore, even in patients with extracranial CMs or AVMs, performing diagnostic evaluations for cerebral AVMs and using measures to prevent intracerebral hemorrhage are crucial. CLINICAL QUESTION/LEVEL OF EVIDENCE Risk, II.
Collapse
Affiliation(s)
- Jeong Yeop Ryu
- From the Departments of Plastic and Reconstructive Surgery
| | | | - Joon Seok Lee
- From the Departments of Plastic and Reconstructive Surgery
| | | | - Jung Dug Yang
- From the Departments of Plastic and Reconstructive Surgery
| | | | | | | | | | - Ho Yun Chung
- From the Departments of Plastic and Reconstructive Surgery
- Cell and Matrix Research Institute, School of Medicine, Kyungpook National University
| |
Collapse
|
10
|
Pereira-Neves A, Dias L, Fragão-Marques M, Vidoedo J, Ribeiro H, Andrade JP, Rocha-Neves J. Monocyte Count as a Predictor of Major Adverse Limb Events in Aortoiliac Revascularization. J Clin Med 2024; 13:6412. [PMID: 39518551 PMCID: PMC11546730 DOI: 10.3390/jcm13216412] [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: 08/30/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: Atherosclerosis is a leading cause of death, especially in the developed world, and is marked by chronic arterial inflammation and lipid accumulation. As key players in its progression, monocytes contribute to plaque formation, inflammation, and tissue repair. Understanding monocyte involvement is crucial for developing better therapeutic approaches. The objective of this study is to assess the prognostic value of monocytes for limb-related outcomes following revascularization for complex aortoiliac lesions, thereby emphasizing the central role of monocytes in atherosclerosis. Methods: This prospective cohort study-enrolled patients who had undergone elective aortoiliac revascularization at two hospitals between January 2013 and December 2023. Patients with TASC II type D lesions were included, excluding those with aneurysmal disease. Demographic, clinical, and procedural data were gathered, and patients were monitored for limb-related outcomes. Preoperative complete blood counts were analyzed, and statistical analyses, including multivariable Cox regression, were conducted to identify predictors of major adverse limb events (MALE). Results: The study included 135 patients with a mean age of 62.4 ± 9.20 years and predominantly male (93%). Patients were followed for a median of 61 IQR [55.4-66.6] months. Smoking history (91%) was the most prevalent cardiovascular risk factor. Preoperative monocyte count >0.720 × 109/L was associated with worse 30-day limb-related outcomes (MALE: OR 7.138 95% CI: 1.509-33.764, p = 0.013) and long-term outcomes, including secondary patency (p = 0.03), major amputation (p = 0.04), and MALE (p = 0.039). Cox regression analysis confirmed an elevated monocyte count as an independent predictor of MALE (adjusted hazard ratio 2.149, 95% CI: 1.115-4.144, p = 0.022). Conclusions: This study demonstrated that patients with a higher absolute monocyte count may be more exposed to the risk of MALE in patients with aortoiliac TASC II type D lesions undergoing revascularization, with predictive accuracy in both the short and long term. Additionally, it was an independent predictor of major amputation. This new marker has the potential to serve as a cost-effective and easily available tool for risk stratification, helping identify patients at higher risk of MALE.
Collapse
Affiliation(s)
- António Pereira-Neves
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, 4050-513 Porto, Portugal; (J.P.A.); (J.R.-N.)
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde de São João, 4200-319 Porto, Portugal;
| | - Lara Dias
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde de São João, 4200-319 Porto, Portugal;
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4050-513 Porto, Portugal
| | - Mariana Fragão-Marques
- Cardiovascular R&D Unit, Faculty of Medicine, University of Porto, 4050-513 Porto, Portugal;
| | - José Vidoedo
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde entre o Tâmega e o Sousa, 4560-136 Penafiel, Portugal;
| | - Hugo Ribeiro
- Community Palliative Care Support Team Gaia, 4430-043 Vila Nova de Gaia, Portugal;
- Faculty of Medicine, University of Coimbra, 3004-535 Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal
- MEDCIDS—Faculty of Medicine, University of Porto, 4050-513 Porto, Portugal
| | - José Paulo Andrade
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, 4050-513 Porto, Portugal; (J.P.A.); (J.R.-N.)
- Rise@Health, Rua Dr. Plácido da Costa, s/n, 4200-450 Porto, Portugal
| | - João Rocha-Neves
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, 4050-513 Porto, Portugal; (J.P.A.); (J.R.-N.)
- Rise@Health, Rua Dr. Plácido da Costa, s/n, 4200-450 Porto, Portugal
| |
Collapse
|
11
|
Athmuri DN, Bhattacharyya J, Bhatnagar N, Shiekh PA. Alleviating hypoxia and oxidative stress for treatment of cardiovascular diseases: a biomaterials perspective. J Mater Chem B 2024; 12:10490-10515. [PMID: 39302443 DOI: 10.1039/d4tb01126k] [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: 09/22/2024]
Abstract
A state of hypoxia (lack of oxygen) persists in the initial and later phases of healing in cardiovascular diseases, which can alter the tissue's repair or regeneration, ultimately affecting the structure and functionality of the related organ. Consequently, this results in a cascade of events, leading to metabolic stress and the production of reactive oxygen species (ROS) and autophagy. This unwanted situation not only limits the oxygen supply to the needy tissues but also creates an inflammatory state, limiting the exchange of nutrients and other supplements. Consequently, biomaterials have gained considerable attention to alleviate hypoxia and oxidative stress in cardiovascular diseases. Numerous oxygen releasing and antioxidant biomaterials have been developed and proven to alleviate hypoxia and oxidative stress. This review article summarizes the mechanisms involved in cardiovascular pathologies due to hypoxia and oxidative stress, as well as the treatment modalities currently in practice. The applications, benefits and possible shortcomings of these approaches have been discussed. Additionally, the review explores the role of novel biomaterials in combating the limitations of existing approaches, primarily focusing on the development of oxygen-releasing and antioxidant biomaterials for cardiac repair and regeneration. It also directs attention to various other potential applications with critical insights for further advancement in this area.
Collapse
Affiliation(s)
- Durga Nandini Athmuri
- SMART Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India.
| | - Jayanta Bhattacharyya
- Bio-therapeutics Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India
| | - Naresh Bhatnagar
- Department of Mechanical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India
| | - Parvaiz Ahmad Shiekh
- SMART Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India.
| |
Collapse
|
12
|
Chen R, Duffy Á, Petrazzini BO, Vy HM, Stein D, Mort M, Park JK, Schlessinger A, Itan Y, Cooper DN, Jordan DM, Rocheleau G, Do R. Expanding drug targets for 112 chronic diseases using a machine learning-assisted genetic priority score. Nat Commun 2024; 15:8891. [PMID: 39406732 PMCID: PMC11480483 DOI: 10.1038/s41467-024-53333-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Identifying genetic drivers of chronic diseases is necessary for drug discovery. Here, we develop a machine learning-assisted genetic priority score, which we call ML-GPS, that incorporates genetic associations with predicted disease phenotypes to enhance target discovery. First, we construct gradient boosting models to predict 112 chronic disease phecodes in the UK Biobank and analyze associations of predicted and observed phenotypes with common, rare, and ultra-rare variants to model the allelic series. We integrate these associations with existing evidence using gradient boosting with continuous feature encoding to construct ML-GPS, training it to predict drug indications in Open Targets and externally testing it in SIDER. We then generate ML-GPS predictions for 2,362,636 gene-phecode pairs. We find that the use of predicted phenotypes, which identify substantially more genetic associations than observed phenotypes across the allele frequency spectrum, significantly improves the performance of ML-GPS. ML-GPS increases coverage of drug targets, with the top 1% of all scores providing support for 15,077 gene-phecode pairs that previously had no support. ML-GPS can also identify well-known target-disease relationships, promising targets without indicated drugs, and targets for several drugs in clinical trials, including LRRK2 inhibitors for Parkinson's disease and olpasiran for cardiovascular disease.
Collapse
Affiliation(s)
- Robert Chen
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Áine Duffy
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ben O Petrazzini
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ha My Vy
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David Stein
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew Mort
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Joshua K Park
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuval Itan
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David N Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Daniel M Jordan
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ghislain Rocheleau
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
13
|
Patel S, Shah N, D’Mello B, Lee A, Werstuck GH. Myeloid GSK3α Deficiency Reduces Lesional Inflammation and Neovascularization during Atherosclerotic Progression. Int J Mol Sci 2024; 25:10897. [PMID: 39456687 PMCID: PMC11507289 DOI: 10.3390/ijms252010897] [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: 09/03/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
The molecular mechanisms by which cardiovascular risk factors promote the development of atherosclerosis are poorly understood. We have recently shown that genetic ablation of myeloid glycogen synthase kinase (GSK)-3α attenuates atherosclerotic lesion development in low-density lipoprotein receptor-deficient (Ldlr-/-) mice. However, the precise contributions of GSK3α/β in atherogenesis are not known. The aim of this study is to investigate the effect of GSK3α and/or β deficiency on lesional inflammation and plaque vascularization. Five-week-old female Ldlr-/- mice were fed a high-fat diet for 10 weeks to establish atherosclerotic lesions. Mice were harvested at 15 weeks of age and atherosclerotic lesions were characterized. The results indicate that, in addition to significantly reducing plaque volume, GSK3α-deficiency decreases inflammation, reduces vasa vasorum density at the aortic sinus, and reduces plasma c-reactive protein (CRP) levels. GSK3β-deficiency is associated with decreased plasma CRP levels but does not affect lesional inflammation or vascularization. These results suggest GSK3α may be an applicable target for the development of novel anti-atherogenic therapies.
Collapse
Affiliation(s)
- Sarvatit Patel
- The Thrombosis and Atherosclerosis Research Institute, Hamilton, ON L8L 2X2, Canada; (S.P.); (N.S.); (A.L.)
| | - Nisarg Shah
- The Thrombosis and Atherosclerosis Research Institute, Hamilton, ON L8L 2X2, Canada; (S.P.); (N.S.); (A.L.)
| | - Brooke D’Mello
- The Thrombosis and Atherosclerosis Research Institute, Hamilton, ON L8L 2X2, Canada; (S.P.); (N.S.); (A.L.)
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Anson Lee
- The Thrombosis and Atherosclerosis Research Institute, Hamilton, ON L8L 2X2, Canada; (S.P.); (N.S.); (A.L.)
| | - Geoff H. Werstuck
- The Thrombosis and Atherosclerosis Research Institute, Hamilton, ON L8L 2X2, Canada; (S.P.); (N.S.); (A.L.)
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
| |
Collapse
|
14
|
Zhao M, Feng L, Li W. Network Pharmacology and Experimental Verification: SanQi-DanShen Treats Coronary Heart Disease by Inhibiting the PI3K/AKT Signaling Pathway. Drug Des Devel Ther 2024; 18:4529-4550. [PMID: 39399124 PMCID: PMC11471080 DOI: 10.2147/dddt.s480248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/11/2024] [Indexed: 10/15/2024] Open
Abstract
Objective To employee network pharmacology to predict the components and pathways of SanQi-DanShen (SQDS) in treating coronary heart disease, followed by in vitro experiments to validate the molecular mechanism of SQDS in treating coronary heart disease. Methods We sourced the active ingredients and targets of Panax notoginseng and Danshen from the Traditional Chinese Medicine Systems Pharmacology database. Coronary heart disease related genes were retrieved from the OMIM, Genecards, and Therapeutic Target databases. Using Cytoscape 3.7.2 software, we constructed a network diagram illustrating the components and targets of SQDS. The associated targets were then imported into the STRING database to build a protein-protein interaction network. The Metascape database and WeChat software were utilized for Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses. Lastly, we performed molecular docking between the key components and related targets using AutoDock Vina. To validate the potential mechanism of SQDS in treating coronary heart disease, we established an acute coronary heart disease rat model via tail vein injection of pituitrin. Results Network pharmacology analysis revealed that 65 active ingredients and 167 targets of SQDS are implicated in the treatment of coronary heart disease. The key targets identified include AKT1, TNF, TP53, IL6, and VEGFA. Notably, the PI3K/AKT signaling pathway emerged as the primary pathway. Furthermore, animal experiments showed that, compared to the model group, SQDS significantly reduced levels of TNF-α, IL-6, Bax, and cardiac troponin I, while increasing Bcl-2 content. It also notably suppressed the expression of p-PI3K and p-AKT, thereby offering protection to myocardial tissue. Conclusion Through the integrated approach of network pharmacology and molecular docking, we have established that SQDS exerts a multi-component, multi-target, and multi-pathway synergistic therapeutic effect on coronary heart disease. Its mechanism may involve the inhibition of the PI3K/AKT signaling pathway and the reduction of inflammatory factor expression.
Collapse
Affiliation(s)
- Min Zhao
- School of Medicine, Lijiang University of Culture and Tourism, Lijiang, Yunnan, 674100, People’s Republic of China
| | - Liuxiang Feng
- People’s Hospital of Yulong Naxi Autonomous County of Lijiang City, Lijiang, Yunnan, 674112, People’s Republic of China
| | - Wenhua Li
- School of Medicine, Xizang Minzu University, Xianyang Shaanxi, 712082, People’s Republic of China
| |
Collapse
|
15
|
Wehbe Z, Wehbe M, Al Khatib A, Dakroub AH, Pintus G, Kobeissy F, Eid AH. Emerging understandings of the role of exosomes in atherosclerosis. J Cell Physiol 2024:e31454. [PMID: 39370679 DOI: 10.1002/jcp.31454] [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: 01/23/2024] [Revised: 08/20/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024]
Abstract
Atherosclerosis remains a major contributor to cardiovascular disease, the leading cause of global morbidity and mortality. Despite the elucidation of several molecular, biochemical, and cellular aspects that contribute to the etio-pathogenesis of atherosclerosis, much remains to be understood about the onset and progression of this disease. Emerging evidence supports a role for exosomes in the cellular basis of atherosclerosis. Indeed, exosomes of activated monocytes seem to accentuate a positive feedback loop that promotes recruitment of pro-inflammatory leukocytes. Moreover, in addition to their role in promoting proliferation and invasion of vascular smooth muscle cells, exosomes can also induce neovascularization within lesions and increase endothelial permeability, two important features of fibrous plaques. Depending on their sources and cargo, exosomes can also induce clot formation and contribute to other hallmarks of atherosclerosis. Taken together, it is becoming increasingly evident that a better understanding of exosome biology is integral to elucidating the pathogenesis of atherosclerosis, and may thus provide insight into a potentially new therapeutic target for this disease.
Collapse
Affiliation(s)
- Zena Wehbe
- Vascular Biology Research Centre, Molecular and Clinical Research Institute, St. George's University of London, London, United Kingdom
| | - Maya Wehbe
- Oxford University Hospitals, Oxford, United Kingdom
| | - Ali Al Khatib
- Department of Nutrition and Food Sciences, Lebanese International University, Beirut, Lebanon
| | - Ali H Dakroub
- Departments of Medicine (Cardiology) and Population Health Science and Policy, Blavatnik Family Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro, Sassari, 07100, Italy
| | - Firas Kobeissy
- Department of Neurobiology, Morehouse School of Medicine, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Atlanta, GA, USA
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, P.O. Box 2713, Qatar
| |
Collapse
|
16
|
Pi H, Wang G, Wang Y, Zhang M, He Q, Zheng X, Yin K, Zhao G, Jiang T. Immunological perspectives on atherosclerotic plaque formation and progression. Front Immunol 2024; 15:1437821. [PMID: 39399488 PMCID: PMC11466832 DOI: 10.3389/fimmu.2024.1437821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Atherosclerosis serves as the primary catalyst for numerous cardiovascular diseases. Growing evidence suggests that the immune response is involved in every stage of atherosclerotic plaque evolution. Rapid, but not specific, innate immune arms, including neutrophils, monocytes/macrophages, dendritic cells (DCs) and other innate immune cells, as well as pattern-recognition receptors and various inflammatory mediators, contribute to atherogenesis. The specific adaptive immune response, governed by T cells and B cells, antibodies, and immunomodulatory cytokines potently regulates disease activity and progression. In the inflammatory microenvironment, the heterogeneity of leukocyte subpopulations plays a very important regulatory role in plaque evolution. With advances in experimental techniques, the fine mechanisms of immune system involvement in atherosclerotic plaque evolution are becoming known. In this review, we examine the critical immune responses involved in atherosclerotic plaque evolution, in particular, looking at atherosclerosis from the perspective of evolutionary immunobiology. A comprehensive understanding of the interplay between plaque evolution and plaque immunity provides clues for strategically combating atherosclerosis.
Collapse
Affiliation(s)
- Hui Pi
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Guangliang Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Yu Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ming Zhang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Qin He
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Xilong Zheng
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Guojun Zhao
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ting Jiang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| |
Collapse
|
17
|
Mangoni AA, Zinellu A. The vascular endothelial growth factor as a candidate biomarker of systemic lupus erythematosus: a GRADE-assessed systematic review and meta-analysis. Clin Exp Med 2024; 24:218. [PMID: 39259392 PMCID: PMC11390800 DOI: 10.1007/s10238-024-01487-w] [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: 06/26/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
There is an ongoing search for novel biomarkers of endothelial damage, active disease, and organ dysfunction in systemic lupus erythematosus (SLE). We investigated the role of the vascular endothelial growth factor (VEGF) as a candidate biomarker by conducting a systematic review and meta-analysis of studies examining VEGF concentrations in SLE patients and healthy controls. We searched electronic databases (PubMed, Scopus, and Web of Science) from inception to 31 May 2024 (inclusion criteria: VEGF measurement in SLE patients and healthy controls and SLE patients with and without active disease or specific organ dysfunction in case-control studies, recruitment of adult participants, and availability of the full text in the English language; exclusion criteria: non-case-control studies, participants under 18 years, articles reporting duplicate or irrelevant data, and animal studies). We assessed the risk of bias and the certainty of evidence using the JBI Critical Appraisal Checklist and GRADE, respectively (PROSPERO registration number: CRD42024561636). Circulating VEGF concentrations were significantly higher in SLE patients than in controls (22 studies; standardised mean difference, SMD = 0.71, 95% CI 0.44 to 0.98, p < 0.001; low certainty of evidence). In SLE patients, VEGF concentrations were significantly higher in those with active disease (six studies; SMD = 1.10, 95% CI 0.27 to 1.92, p = 0.009; very low certainty of evidence) and lupus nephritis (four studies; SMD = 0.80, 95% CI 0.03 to 1.57, p = 0.042; very low certainty of evidence). Only one study reported VEGF concentrations in SLE patients with and without pulmonary arterial hypertension. The effect size of the differences in VEGF concentrations between SLE patients and controls was not associated with disease duration, use of glucocorticoids and immunosuppressors, biological matrix assessed, or analytical method used. However, it was significantly associated with the study's geographical location. The evidence was limited by the high but partially explainable heterogeneity and the presence of publication bias which was addressed with the "trim-and-fill" method (SLE presence), the high but partially explainable heterogeneity and lack of assessment of publication bias because of the limited study number (active disease), and the limited study number preventing the identification of sources of heterogeneity, sensitivity analysis, and assessment of publication bias (lupus nephritis). Our results highlight VEGF's potential role as a SLE biomarker and the need for further research, also given the aforementioned limitations, investigating VEGF concentrations in a wide range of SLE patient subgroups.
Collapse
Affiliation(s)
- Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia.
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia.
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, SA, 5042, Australia.
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
18
|
Kirdeev A, Burkin K, Vorobev A, Zbirovskaya E, Lifshits G, Nikolaev K, Zelenskaya E, Donnikov M, Kovalenko L, Urvantseva I, Poptsova M. Machine learning models for predicting risks of MACEs for myocardial infarction patients with different VEGFR2 genotypes. Front Med (Lausanne) 2024; 11:1452239. [PMID: 39301488 PMCID: PMC11410707 DOI: 10.3389/fmed.2024.1452239] [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: 06/20/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024] Open
Abstract
Background The development of prognostic models for the identification of high-risk myocardial infarction (MI) patients is a crucial step toward personalized medicine. Genetic factors are known to be associated with an increased risk of cardiovascular diseases; however, little is known about whether they can be used to predict major adverse cardiac events (MACEs) for MI patients. This study aimed to build a machine learning (ML) model to predict MACEs in MI patients based on clinical, imaging, laboratory, and genetic features and to assess the influence of genetics on the prognostic power of the model. Methods We analyzed the data from 218 MI patients admitted to the emergency department at the Surgut District Center for Diagnostics and Cardiovascular Surgery, Russia. Upon admission, standard clinical measurements and imaging data were collected for each patient. Additionally, patients were genotyped for VEGFR-2 variation rs2305948 (C/C, C/T, T/T genotypes with T being the minor risk allele). The study included a 9-year follow-up period during which major ischemic events were recorded. We trained and evaluated various ML models, including Gradient Boosting, Random Forest, Logistic Regression, and AutoML. For feature importance analysis, we applied the sequential feature selection (SFS) and Shapley's scheme of additive explanation (SHAP) methods. Results The CatBoost algorithm, with features selected using the SFS method, showed the best performance on the test cohort, achieving a ROC AUC of 0.813. Feature importance analysis identified the dose of statins as the most important factor, with the VEGFR-2 genotype among the top 5. The other important features are coronary artery lesions (coronary artery stenoses ≥70%), left ventricular (LV) parameters such as lateral LV wall and LV mass, diabetes, type of revascularization (CABG or PCI), and age. We also showed that contributions are additive and that high risk can be determined by cumulative negative effects from different prognostic factors. Conclusion Our ML-based approach demonstrated that the VEGFR-2 genotype is associated with an increased risk of MACEs in MI patients. However, the risk can be significantly reduced by high-dose statins and positive factors such as the absence of coronary artery lesions, absence of diabetes, and younger age.
Collapse
Affiliation(s)
- Alexander Kirdeev
- Faculty of Computer Science, AI and Digital Science Institute, International Laboratory of Bioinformatics, Higher School of Economics University, Moscow, Russia
| | - Konstantin Burkin
- Faculty of Computer Science, AI and Digital Science Institute, International Laboratory of Bioinformatics, Higher School of Economics University, Moscow, Russia
| | - Anton Vorobev
- Department of Cardiology, Surgut State University, Surgut, Russia
| | - Elena Zbirovskaya
- Faculty of Computer Science, AI and Digital Science Institute, International Laboratory of Bioinformatics, Higher School of Economics University, Moscow, Russia
| | - Galina Lifshits
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| | - Konstantin Nikolaev
- Federal Research Center Institute of Cytology and Genetics, Novosibirsk, Russia
| | - Elena Zelenskaya
- Department of Cardiology, Surgut State University, Surgut, Russia
| | - Maxim Donnikov
- Department of Cardiology, Surgut State University, Surgut, Russia
| | - Lyudmila Kovalenko
- Department of General Pathology and Pathophysiology, Surgut State University, Surgut, Russia
| | - Irina Urvantseva
- Department of Cardiology, Surgut State University, Surgut, Russia
- Ugra Center for Diagnostics and Cardiovascular Surgery, Surgut, Russia
| | - Maria Poptsova
- Faculty of Computer Science, AI and Digital Science Institute, International Laboratory of Bioinformatics, Higher School of Economics University, Moscow, Russia
| |
Collapse
|
19
|
Ugusman A, Hisam NSN, Othman NS, Anuar NNM, Hamid AA, Kumar J, Razmi MM, Aminuddin A. Pharmacological interventions for intraplaque neovascularization in atherosclerosis. Pharmacol Ther 2024; 261:108685. [PMID: 38977083 DOI: 10.1016/j.pharmthera.2024.108685] [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: 03/02/2024] [Revised: 06/29/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Advanced atherosclerosis is linked to plaque instability, which can result in rupture and the onset of a heart attack. Evidence gathered from human atheroma plaques indicates that intraplaque neovascularization poses a risk to plaque stability and may lead to plaque hemorrhage. Hence, targeting the neovascularization within the atheroma plaque has the potential to mitigate the plaque's vulnerability. While neovascularization has been extensively explored in the context of cancer, research on pharmacological inhibition of this phenomenon in atherosclerosis remains limited. This systematic review aimed to comprehensively assess current and emerging pharmacological interventions for inhibiting intraplaque neovascularization in preclinical settings. Electronic databases (Web of Science, PubMed, Scopus, and Ovid) were searched from January 2013 until February 1, 2024. Preclinical studies reporting the effect of any pharmacological interventions targeting intraplaque neovascularization were included. A total of 10 articles involving in vivo animal studies were eligible for inclusion, with five of them incorporating in vitro experiments to complement their in vivo findings. The pharmacological interventions studied were axitinib, ghrelin, K5, rosuvastatin, atorvastatin, 3PO, everolimus, melatonin, Si-Miao-Yong-A, and protocatechuic aldehyde. All the interventions showed a positive impact in inhibiting intraplaque neovascularization in various atherosclerotic animal models through various signaling pathways. This review provides valuable insights into pharmacological approaches to attenuate intraplaque neovascularization that could serve as a promising therapeutic avenue to enhance plaque stability.
Collapse
Affiliation(s)
- Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia.
| | - Nur Syahidah Nor Hisam
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia; Programme of Biomedical Science, Centre for Toxicology & Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Nur Syakirah Othman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Nur Najmi Mohamad Anuar
- Programme of Biomedical Science, Centre for Toxicology & Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Adila A Hamid
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Maisarah Md Razmi
- Programme of Biomedical Science, Centre for Toxicology & Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Amilia Aminuddin
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia.
| |
Collapse
|
20
|
Lee SM, Yoon BH, Lee JW, Jeong IJY, Kim I, Pack CG, Kim YH, Ha CH. Circulating miRNA-4701-3p as a predictive biomarker of cardiovascular disease which induces angiogenesis by inhibition of TOB2. Microvasc Res 2024; 155:104698. [PMID: 38801943 DOI: 10.1016/j.mvr.2024.104698] [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: 08/24/2023] [Revised: 05/07/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Angiogenesis is mainly regulated by the delivery of VEGF-dependent signaling to cells. However, the angiogenesis mechanism regulated by VEGF-induced miRNA is still not understood. After VEGF treatment in HUVECs, we screened the changed miRNAs through small-RNA sequencing and found VEGF-induced miR-4701-3p. Furthermore, the GFP reporter gene was used to reveal that TOB2 expression was regulated by miR-4701-3p, and it was found that TOB2 and miR-4701-3p modulation could cause angiogenesis in an in-vitro angiogenic assay. Through the luciferase assay, it was confirmed that the activation of the angiogenic transcription factor MEF2 was regulated by the suppression and overexpression of TOB2 and miR-4701-3p. As a result, MEF2 downstream gene mRNAs that induce angiogenic function were regulated. We used the NCBI GEO datasets to reveal that the expression of TOB2 and MEF2 was significantly changed in cardiovascular disease. Finally, it was confirmed that the expression of circulating miR-4701-3p in the blood of myocardial infarction patients was remarkably increased. In patients with myocardial infarction, circulating miR-4701-3p was increased regardless of age, BMI, and sex, and showed high AUC levels in specificity and sensitivity analysis (AUROC) (AUC = 0.8451, 95 % CI 0.78-0.90). Our data showed TOB2-mediated modulation of MEF2 and its angiogenesis by VEGF-induced miR-4701-3p in vascular endothelial cells. In addition, through bioinformatics analysis using GEO data, changes in TOB2 and MEF2 were revealed in cardiovascular disease. We suggest that circulating miR-4701-3p has high potential as a biomarker for myocardial infarction.
Collapse
Affiliation(s)
- Seung Min Lee
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Bo Hyun Yoon
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Woo Lee
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - I Jin-Yong Jeong
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Inki Kim
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; ConveRgence mEDIcine research cenTer (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Chan-Gi Pack
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; ConveRgence mEDIcine research cenTer (CREDIT), Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Young-Hak Kim
- Cardiology Division, Asan Medical Center and University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Chang Hoon Ha
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
21
|
Correia AF, Oliveira CGCD, Oliveira DCD, Pereira MC, Carvalho FA, Martins ECC, Oliveira DCD. Circulating Interleukin-22 in Patients with Acute Myocardial Infarction Undergoing Primary Percutaneous Coronary Intervention. J Clin Med 2024; 13:4971. [PMID: 39274184 PMCID: PMC11396034 DOI: 10.3390/jcm13174971] [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: 07/02/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/16/2024] Open
Abstract
Acute coronary syndrome (ACS) represents an important clinical manifestation of coronary artery disease (CAD) and is characterized by a particularly poor prognosis. Myocardial reperfusion through primary percutaneous coronary intervention (PPCI) is imperative in the event of acute ST elevation myocardial infarction (STEMI). Interleukin-22 (IL-22) regulates immune and inflammatory responses. This interleukin has been described in the scenario of the CAD, but there are no data in patients with STEMI undergoing PPCI. Objectives: The goals of this study were to investigate the differences in circulating IL-22 levels between patients with STEMI undergoing PPCI and healthy controls and to determine whether these differences were associated with the culprit coronary artery, door-to-balloon time (DBT), final angiographic result, CAD classification, and presence of diabetes mellitus (DM). Methods: A total of 280 participants were recruited, comprising 210 STEMI cases and 70 healthy controls. Participants underwent clinical and angiographic evaluations, and serum IL-22 levels were measured using an enzyme-linked immunosorbent assay (ELISA). Data analysis was performed using the Mann-Whitney and Fisher tests, with p < 0.05 indicating significance. Results: Serum IL-22 levels were lower in cases (149.63, 84.99-294.56) than in the controls (482.67, 344.33-641.00); p < 0.001. Lower IL-22 levels were associated with the right coronary artery (RCA) (144.57, 70.84-242.43; 146.00, 63.60-279.67; 191.71, 121.80-388.97); p = 0.033. IL-22 was lower with shorter DBT (≤60 min, 106.00, 49.60-171.71; >60 min, 153.00, 88.86-313.60); p = 0.043. Conclusions: IL-22 levels were significantly lower in patients with STEMI than in healthy controls.
Collapse
Affiliation(s)
- Augusto Ferreira Correia
- Internal Medicine Department, Cardiology Division, Federal University of Pernambuco, Recife 50670-901, Brazil
- Cardiology Emergency Room of Pernambuco, University of Pernambuco, Recife 52010-010, Brazil
| | | | | | - Michelly Cristina Pereira
- Internal Medicine Department, Cardiology Division, Federal University of Pernambuco, Recife 50670-901, Brazil
| | | | | | - Dinaldo Cavalcanti de Oliveira
- Internal Medicine Department, Cardiology Division, Federal University of Pernambuco, Recife 50670-901, Brazil
- Cardiology Emergency Room of Pernambuco, University of Pernambuco, Recife 52010-010, Brazil
| |
Collapse
|
22
|
Luo J, Wang L, Cui C, Chen H, Zeng W, Li X. MicroRNA-19a-3p inhibits endothelial dysfunction in atherosclerosis by targeting JCAD. BMC Cardiovasc Disord 2024; 24:394. [PMID: 39080547 PMCID: PMC11287888 DOI: 10.1186/s12872-024-04063-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/19/2024] [Indexed: 08/03/2024] Open
Abstract
OBJECTIVE To examine the influences and mechanisms of MicroRNA-19a-3p (miR-19a-3p) on endothelial dysfunction in atherosclerosis. METHODS An analysis of miR-19a expression was carried out using the Gene Expression Omnibus (GEO) database. The effect of miR-19a-3p on endothelial function in HUVECs was evaluated by miR-19a-3p overexpression under TNF-α treatment. Luciferase assays were performed to explore the potential target genes. Overexpression of junctional protein associated with coronary artery disease (JCAD) was used to examine the effects of miR-19a-3p on cell adhesion, and proliferation. RESULTS MiR-19a-3p expression in endothelial cells decreased after exposure to TNF-α and/or oscillatory flow, consistent with the expression change of miR-19a-3p found in atherosclerotic plaques. Additionally, endothelial cell dysfunction and inflammation were significantly diminished by miR-19a-3p overexpression but markedly exacerbated by miR-19a-3p inhibition. MiR-19a-3p transfection significantly decreased the expression of JCAD by binding to the 3'-UTR of JCAD mRNA. Furthermore, the protective effect of miR-19a-3p against endothelial cell dysfunction and inflammation was achieved by regulating JCAD and was closely linked to the Hippo/YAP signaling pathway. CONCLUSION MiR-19a-3p expression is a crucial molecular switch in the onset of atherosclerosis and miR-19a-3p overexpression is a possible pharmacological therapeutic strategy for reversing the development of atherosclerosis.
Collapse
Affiliation(s)
- Jinque Luo
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, "The 14th Five-Year Plan" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), College of Pharmacy, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China
- College of Pharmacy, Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, 410219, Hunan, China
| | - Ling Wang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, "The 14th Five-Year Plan" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), College of Pharmacy, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China
- College of Pharmacy, Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, 410219, Hunan, China
| | - Chaoyue Cui
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, "The 14th Five-Year Plan" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), College of Pharmacy, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China
| | - Hongyu Chen
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, "The 14th Five-Year Plan" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), College of Pharmacy, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China
| | - Wanli Zeng
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, "The 14th Five-Year Plan" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), College of Pharmacy, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China
| | - Xin Li
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, "The 14th Five-Year Plan" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), College of Pharmacy, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China.
| |
Collapse
|
23
|
Zhou L, Wu J, Wei Z, Zheng Y. Legumain in cardiovascular diseases. Exp Biol Med (Maywood) 2024; 249:10121. [PMID: 39104790 PMCID: PMC11298360 DOI: 10.3389/ebm.2024.10121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide, having become a global public health problem, so the pathophysiological mechanisms and therapeutic strategies of CVDs need further study. Legumain is a powerful enzyme that is widely distributed in mammals and plays an important role in a variety of biological processes. Recent research suggests that legumain is associated with the occurrence and progression of CVDs. In this review, we provide a comprehensive overview of legumain in the pathogenesis of CVDs. The role of legumain in CVDs, such as carotid atherosclerosis, pulmonary hypertension, coronary artery disease, peripheral arterial disease, aortic aneurysms and dissection, is discussed. The potential applications of legumain as a biomarker of these diseases are also explored. By understanding the role of legumain in the pathogenesis of CVDs, we aim to support new therapeutic strategies to prevent or treat these diseases.
Collapse
Affiliation(s)
- Lei Zhou
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Vascular Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianqiang Wu
- Institute of Clinical Medicine, National Science and Technology Key Infrastructure on Translational Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zairong Wei
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yuehong Zheng
- Department of Vascular Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
24
|
Zheng M, Liu Y, Zhang G, Yang Z, Xu W, Chen Q. The Antioxidant Properties, Metabolism, Application and Mechanism of Ferulic Acid in Medicine, Food, Cosmetics, Livestock and Poultry. Antioxidants (Basel) 2024; 13:853. [PMID: 39061921 PMCID: PMC11273498 DOI: 10.3390/antiox13070853] [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: 05/27/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Ferulic acid is a ubiquitous ingredient in cereals, vegetables, fruits and Chinese herbal medicines. Due to the ferulic phenolic nucleus coupled to an extended side chain, it readily forms a resonant-stable phenoxy radical, which explains its potent antioxidant potential. In addition, it also plays an important role in anti-cancer, pro-angiogenesis, anti-thrombosis, neuroprotection, food preservation, anti-aging, and improving the antioxidant performance of livestock and poultry. This review provides a comprehensive summary of the structure, mechanism of antioxidation, application status, molecular mechanism of pharmacological activity, existing problems, and application prospects of ferulic acid and its derivatives. The aim is to establish a theoretical foundation for the utilization of ferulic acid in medicine, food, cosmetics, livestock, and poultry.
Collapse
Affiliation(s)
| | | | | | | | | | - Qinghua Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
25
|
Zhang J, Han Y, Jia R, Zhu Q, Wang X, Liu M, Zhang W. Exploring the role of myeloperoxidase in the atherosclerotic process in hypoxic mice based on the MAPK signaling pathway. Biochem Pharmacol 2024; 225:116275. [PMID: 38729447 DOI: 10.1016/j.bcp.2024.116275] [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: 12/24/2023] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
Atherosclerosis (AS) is the common pathophysiological basis of various cardiovascular diseases and the leading cause of death from cardiovascular disease worldwide. When the body is in a hypoxic environment, enhanced oxidative stress and significant accumulation of reactive oxygen species (ROS) in tissue cells exacerbate the inflammatory response, resulting in increased release of myeloperoxidase (MPO), catalyzing the formation of large quantities of hypochlorous acid (HOCl), further oxidative modification of low-density lipoprotein (LDL), and exacerbating the formation and progression of atherosclerotic plaques. The MAPK signaling pathway is important in oxidative stress-mediated promotion of atherogenesis. MPO -/- mice were used in this study to establish a hypoxia model simulating 5000 m altitude and a Western high-fat diet-induced atherosclerosis model for 12 weeks. Exploring the role of MPO in the atherosclerotic process in hypoxic mice by observing the MAPK signaling pathway to provide a therapeutic target for the prevention and treatment of hypoxic atherosclerotic disease in the plateau. We found that hypoxia promotes the formation of atherosclerosis in mice, and the mechanism may be that increased MPO in vivo promotes an inflammatory response, which plays a crucial role in the formation of atherosclerosis. In addition, hypoxia further exacerbates plaque instability by activating the MAPK signaling pathway to upregulate vascular endothelial growth factor (VEGF) and matrix metalloproteinase-9 (MMP9), which in turn promotes angiogenesis within the plaque. Therefore, a potential target for preventing and treating hypoxic atherosclerotic disease is the inhibition of MPO.
Collapse
Affiliation(s)
- Jingxuan Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Ying Han
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Ruhan Jia
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Qinfang Zhu
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China; Qinghai Provincial People's Hospital, Xining, Qinghai, China
| | - Xiaozhou Wang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China; Department of Hypertension, Qinghai Cardio-Cerebrovascular Hospital, Xining, Qinghai, China
| | - Meiheng Liu
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Wei Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China.
| |
Collapse
|
26
|
Mohammed KAK, Madeddu P, Avolio E. MEK inhibitors: a promising targeted therapy for cardiovascular disease. Front Cardiovasc Med 2024; 11:1404253. [PMID: 39011492 PMCID: PMC11247000 DOI: 10.3389/fcvm.2024.1404253] [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: 03/20/2024] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Cardiovascular disease (CVD) represents the leading cause of mortality and disability all over the world. Identifying new targeted therapeutic approaches has become a priority of biomedical research to improve patient outcomes and quality of life. The RAS-RAF-MEK (mitogen-activated protein kinase kinase)-ERK (extracellular signal-regulated kinase) pathway is gaining growing interest as a potential signaling cascade implicated in the pathogenesis of CVD. This pathway is pivotal in regulating cellular processes like proliferation, growth, migration, differentiation, and survival, which are vital in maintaining cardiovascular homeostasis. In addition, ERK signaling is involved in controlling angiogenesis, vascular tone, myocardial contractility, and oxidative stress. Dysregulation of this signaling cascade has been linked to cell dysfunction and vascular and cardiac pathological remodeling, which contribute to the onset and progression of CVD. Recent and ongoing research has provided insights into potential therapeutic interventions targeting the RAS-RAF-MEK-ERK pathway to improve cardiovascular pathologies. Preclinical studies have demonstrated the efficacy of targeted therapy with MEK inhibitors (MEKI) in attenuating ERK activation and mitigating CVD progression in animal models. In this article, we first describe how ERK signaling contributes to preserving cardiovascular health. We then summarize current knowledge of the roles played by ERK in the development and progression of cardiac and vascular disorders, including atherosclerosis, myocardial infarction, cardiac hypertrophy, heart failure, and aortic aneurysm. We finally report novel therapeutic strategies for these CVDs encompassing MEKI and discuss advantages, challenges, and future developments for MEKI therapeutics.
Collapse
Affiliation(s)
- Khaled A K Mohammed
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paolo Madeddu
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Elisa Avolio
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
27
|
Liu Y, Jiang Z, Yang X, Wang Y, Yang B, Fu Q. Engineering Nanoplatforms for Theranostics of Atherosclerotic Plaques. Adv Healthc Mater 2024; 13:e2303612. [PMID: 38564883 DOI: 10.1002/adhm.202303612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/28/2024] [Indexed: 04/04/2024]
Abstract
Atherosclerotic plaque formation is considered the primary pathological mechanism underlying atherosclerotic cardiovascular diseases, leading to severe cardiovascular events such as stroke, acute coronary syndromes, and even sudden cardiac death. Early detection and timely intervention of plaques are challenging due to the lack of typical symptoms in the initial stages. Therefore, precise early detection and intervention play a crucial role in risk stratification of atherosclerotic plaques and achieving favorable post-interventional outcomes. The continuously advancing nanoplatforms have demonstrated numerous advantages including high signal-to-noise ratio, enhanced bioavailability, and specific targeting capabilities for imaging agents and therapeutic drugs, enabling effective visualization and management of atherosclerotic plaques. Motivated by these superior properties, various noninvasive imaging modalities for early recognition of plaques in the preliminary stage of atherosclerosis are comprehensively summarized. Additionally, several therapeutic strategies are proposed to enhance the efficacy of treating atherosclerotic plaques. Finally, existing challenges and promising prospects for accelerating clinical translation of nanoplatform-based molecular imaging and therapy for atherosclerotic plaques are discussed. In conclusion, this review provides an insightful perspective on the diagnosis and therapy of atherosclerotic plaques.
Collapse
Affiliation(s)
- Yuying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Zeyu Jiang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Bin Yang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
28
|
Größer V, Weyh C, Böttrich T, Frech T, Nolte S, Sommer N, Huber M, Eder K, Dörr O, Hoelscher S, Weber R, Akdogan E, Nef H, Most A, Hamm CW, Krüger K, Bauer P. Association of cardiorespiratory fitness level with vascular function and subclinical atherosclerosis in the elderly. Eur J Appl Physiol 2024; 124:1487-1497. [PMID: 38133663 PMCID: PMC11055712 DOI: 10.1007/s00421-023-05375-1] [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: 08/22/2023] [Accepted: 11/19/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE Physical exercise is crucial for healthy aging and plays a decisive role in the prevention of atherosclerotic cardiovascular disease (ASCVD). A higher level of cardiorespiratory fitness (CRF) in the elderly is associated with lower cardiovascular and all-cause mortality. This study investigated the association of CRF level with vascular function and cardiovascular risk factors in the elderly. METHODS We examined 79 apparently healthy and physically active subjects aged > 55 years (64 ± 4 years). Cardiovascular functional parameters assessed included brachial and central blood pressure (BP), pulse wave velocity (PWV), augmentation index (Aix), and ankle-brachial index. Sonography of the common carotid artery was performed. CRF level was determined by a cardiopulmonary exercise test, and everyday activity was quantified with an accelerometer. RESULTS All participants had a higher CRF level than the reported age-specific normative values. Twenty-nine subjects had subclinical atherosclerosis of the common carotid artery. Compared with participants without atherosclerosis, they were older (p = 0.007), displayed higher brachial systolic BP (p = 0.006), and higher central systolic BP (p = 0.014). Lower brachial (p = 0.036) and central (p = 0.003) systolic BP, lower PWV (p = 0.004), lower Aix (p < 0.001), lower body fat percentage (< 0.001), and lower LDL cholesterol (p = 0.005) were associated with a higher CRF level. CONCLUSIONS In this cohort of healthy and physically active individuals, subjects with subclinical atherosclerosis displayed higher systolic brachial and central BP. A higher CRF level was associated with enhanced vascular function, consistent with an influence of CRF on both BP and vascular function in the elderly.
Collapse
Affiliation(s)
- Vincent Größer
- Department of Cardiology and Angiology, Justus- Liebig-University Giessen, 35390, Giessen, Germany
| | - Christopher Weyh
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University, Giessen, Germany
| | - Tim Böttrich
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University, Giessen, Germany
| | - Torsten Frech
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University, Giessen, Germany
| | - Svenja Nolte
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University, Giessen, Germany
| | - Natascha Sommer
- Department of Internal Medicine, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Justus-Liebig-University Giessen, Giessen, Germany
| | - Magdalena Huber
- Institute for Systems Immunology, Center for Tumor und Immunology, Marburg, Germany
| | - Klaus Eder
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University, Giessen, Germany
| | - Oliver Dörr
- Department of Cardiology and Angiology, Justus- Liebig-University Giessen, 35390, Giessen, Germany
| | - Sophie Hoelscher
- Department of Cardiology and Angiology, Justus- Liebig-University Giessen, 35390, Giessen, Germany
| | - Rebecca Weber
- Department of Cardiology and Angiology, Justus- Liebig-University Giessen, 35390, Giessen, Germany
| | - Ebru Akdogan
- Department of Cardiology and Angiology, Justus- Liebig-University Giessen, 35390, Giessen, Germany
| | - Holger Nef
- Department of Cardiology and Angiology, Justus- Liebig-University Giessen, 35390, Giessen, Germany
| | - Astrid Most
- Department of Cardiology and Angiology, Justus- Liebig-University Giessen, 35390, Giessen, Germany
| | - Christian W Hamm
- Department of Cardiology and Angiology, Justus- Liebig-University Giessen, 35390, Giessen, Germany
- Department of Cardiology, Kerckhoff Clinic GmbH, Bad Nauheim, Germany
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University, Giessen, Germany
| | - Pascal Bauer
- Department of Cardiology and Angiology, Justus- Liebig-University Giessen, 35390, Giessen, Germany.
| |
Collapse
|
29
|
Zhou Q, Li H, Cheng Y, Ma X, Tang S, Tang C. Pax-8: Molecular biology, pathophysiology, and potential pathogenesis. Biofactors 2024; 50:408-421. [PMID: 37988248 DOI: 10.1002/biof.2016] [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: 03/14/2023] [Accepted: 10/21/2023] [Indexed: 11/23/2023]
Abstract
Transcription factors, as the convergence points of multiple signaling pathways in eukaryotic cells, are closely involved in disease development. Pax-8, an important transcription factor belonging to the Pax family, exerts a crucial influence on the regulation of gene expression required for both physiological conditions and pathological processes. Pax-8 contributes to the pathogenesis of many human diseases, ranging from cardiovascular disease to many cancers, and therefore, it can be imagined that Pax-8 holds great therapeutic potential. In this review, we summarize the structure, distribution, function, and regulatory mechanisms of Pax-8 to provide a new research direction for Pax-8.
Collapse
Affiliation(s)
- Qinyi Zhou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Cardiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Heng Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yaqiong Cheng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaofeng Ma
- Department of Cardiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shilin Tang
- Department of Critical Care Medicine, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chaoke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
30
|
Brandes F, Meidert AS, Kirchner B, Yu M, Gebhardt S, Steinlein OK, Dolch ME, Rantner B, Tsilimparis N, Schelling G, Pfaffl MW, Reithmair M. Identification of microRNA biomarkers simultaneously expressed in circulating extracellular vesicles and atherosclerotic plaques. Front Cardiovasc Med 2024; 11:1307832. [PMID: 38725837 PMCID: PMC11079260 DOI: 10.3389/fcvm.2024.1307832] [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: 10/05/2023] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
Background Atherosclerosis is a widespread disorder of the cardiovascular system. The early detection of plaques by circulating biomarkers is highly clinically relevant to prevent the occurrence of major complications such as stroke or heart attacks. It is known that extracellular vesicles (EVs) are important in intercellular communication in atherosclerotic disorders and carry many components of their cells of origin, including microRNAs (miRNAs). In this study, we test the assumption that miRNAs present in material acquired from plaques in patients undergoing surgery for atherosclerotic carotid artery stenosis are also expressed in circulating EVs obtained from the identical patients. This would allow the adoption of a liquid biopsy approach for the detection of plaques. Methods We studied 22 surgical patients with atherosclerotic carotid arterial stenosis and 28 healthy controls. EVs were isolated from serum by precipitation. miRNA expression profiles of serum-derived EVs were obtained by small RNA sequencing and in plaque material simultaneously acquired from patients. A comparative analysis was performed to identify circulating atherosclerosis-associated miRNAs that are also detectable in plaques. Results Seven miRNAs were found to be differentially regulated in patient serum compared with the serum of healthy controls. Of these, miR-193b-5p, miR-193a-5p, and miR-125a-3p were significantly upregulated in patients compared with that in healthy controls and present in both, circulating EVs and plaque material. An overrepresentation analysis of experimentally validated mRNA targets revealed an increased regulation of inflammation and vascular growth factors, key players in atherosclerosis and plaque formation. Conclusion Our findings suggest that circulating EVs reflect plaque development in patients with symptomatic carotid artery stenosis, which can serve as biomarker candidates for detecting the presence of atherosclerotic plaques.
Collapse
Affiliation(s)
- Florian Brandes
- Department of Anesthesiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Agnes S. Meidert
- Department of Anesthesiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Benedikt Kirchner
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Mia Yu
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Sonja Gebhardt
- Department of Anaesthesiology, InnKlinikum Altötting, Altötting, Germany
| | - Ortrud K. Steinlein
- Institute of Human Genetics, LMU University Hospital, LMU Munich, Munich, Germany
| | - Michael E. Dolch
- Department of Anaesthesiology, InnKlinikum Altötting, Altötting, Germany
| | - Barbara Rantner
- Department of Vascular Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Nikolaos Tsilimparis
- Department of Vascular Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Gustav Schelling
- Department of Anesthesiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Michael W. Pfaffl
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Marlene Reithmair
- Institute of Human Genetics, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
31
|
Lu H, Xu Y, Zhao H, Xu X. A novel rabbit model of atherosclerotic vulnerable plaque established by cryofluid-induced endothelial injury. Sci Rep 2024; 14:9447. [PMID: 38658774 PMCID: PMC11043414 DOI: 10.1038/s41598-024-60287-0] [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: 01/06/2024] [Accepted: 04/21/2024] [Indexed: 04/26/2024] Open
Abstract
Acute thrombosis secondary to atherosclerotic plaque rupture is the main cause of acute cardiac and cerebral ischemia. An animal model of unstable atherosclerotic plaques is highly important for investigating the mechanism of plaque rupture and thrombosis. However, current animal models involve complex operations, are costly, and have plaque morphologies that are different from those of humans. We aimed to establish a simple animal model of vulnerable plaques similar to those of humans. Rabbits were randomly divided into three groups. Group A was given a normal formula diet for 13 weeks. Group C underwent surgery on the intima of the right carotid artery with - 80 °C cryofluid-induced injury after 1 week of a high-fat diet and further feeding a 12-week high-fat diet. Group B underwent the same procedure as Group C but without the - 80 °C cryofluid. Serum lipid levels were detected via ELISA. The plaque morphology, stability and degree of stenosis were evaluated through hematoxylin-eosin (HE) staining, Masson trichrome staining, Elastica van Gieson staining (EVG), and oil red O staining. Macrophages and inflammatory factors in the plaques were assessed via immunohistochemical analysis. The serum low-density lipoprotein cholesterol (LDL-C), triglyceride (TG), and total cholesterol (TC) levels in groups B and C were significantly greater than those in group A. No plaque formation was observed in group A. The plaques in group B were very small. In group C, obvious plaques were observed in the blood vessels, and the plaques exhibited a thin fibrous cap, a large lipid core, and partially visible neovascularization, which is consistent with the characteristics of vulnerable plaques. In the plaques of group C, a large number of macrophages were present, and matrix metalloproteinase 9 (MMP-9) and lectin-like oxidized LDL receptor 1 (LOX-1) were abundantly expressed. We successfully established a rabbit model of vulnerable carotid plaque similar to that of humans through the combination of cryofluid-induced endothelial injury and a high-fat diet, which is feasible and cost effective.
Collapse
Affiliation(s)
- Huaizhi Lu
- Department of Cardiovascular Medicine, First People's Hospital of Shangqiu, Kaixuan South Road 292, Shangqiu, 476000, China.
| | - Yiran Xu
- The Second Naval Hospital of Southern Theater Command of PLA, Sanya, 572029, China
| | - Hui Zhao
- Department of Cardiovascular Medicine, First People's Hospital of Shangqiu, Kaixuan South Road 292, Shangqiu, 476000, China
| | - Xuesheng Xu
- Department of Cardiovascular Medicine, First People's Hospital of Shangqiu, Kaixuan South Road 292, Shangqiu, 476000, China
| |
Collapse
|
32
|
Bai P, Xiang X, Kang J, Xiang X, Jiang J, Fu X, Zhang Y, Li L. DFMG decreases angiogenesis to uphold plaque stability by inhibiting the TLR4/VEGF pathway in mice. PLoS One 2024; 19:e0302387. [PMID: 38635560 PMCID: PMC11025810 DOI: 10.1371/journal.pone.0302387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/28/2024] [Indexed: 04/20/2024] Open
Abstract
The aim of this study was to elucidate the specific mechanism through which 7-difluoromethoxy-5,4'-dimethoxygenistein (DFMG) inhibits angiogenesis in atherosclerosis (AS) plaques, given its previously observed but poorly understood inhibitory effects. In vitro, a model using Human Umbilical Vein Endothelial (HUVEC-12) cells simulated the initial lesion in the atherosclerotic pathological process, specifically oxidative stress injury, by exposing cells to 30 μmol/L LPC. Additionally, an AS mouse model was developed in ApoE knockout mice through a 16-week period of high-fat feeding. DFMG demonstrated a reduction in tubule quantities in the tube formation assay and neovascularization induced by oxidative stress-damaged endothelial cells in the chicken embryo chorioallantoic membrane assay. Furthermore, DFMG decreased lipid levels in the blood of ApoE knockout mice with AS, along with a decrease in atherosclerotic plaques and neovascularizations in the aortic arch and descending aorta of AS animal models. DFMG treatment upregulated microRNA140 (miR-140) expression and suppressed VEGF secretion in HUVEC-12 cells. These effects were counteracted by Toll-like receptor 4 (TLR4) overexpression in HUVEC-12 cells subjected to oxidative injury or in a mouse model of AS. Dual-luciferase reporter assays demonstrated that miR-140 directly targeted TLR4. Immunohistochemical assay findings indicated a significant inverse relationship between miR-140 expression and TLR4 expression in ApoE knockout mice subjected to a high-fat diet. The study observed a close association between DFMG inhibitory effects on angiogenesis and plaque stability in AS, and the inhibition of the TLR4/NF-κB/VEGF signaling pathway, negatively regulated by miR-140.
Collapse
Affiliation(s)
- Pingjuan Bai
- Pathology Department, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
- Medical College of Hunan Normal University, Changsha, Hunan Province, China
| | - Xueping Xiang
- Medical College of Hunan Normal University, Changsha, Hunan Province, China
| | - Jiawen Kang
- Medical College of Hunan Normal University, Changsha, Hunan Province, China
| | - Xiaoqing Xiang
- Medical College of Hunan Normal University, Changsha, Hunan Province, China
| | - Jingwen Jiang
- Medical College of Hunan Normal University, Changsha, Hunan Province, China
| | - Xiaohua Fu
- Medical College of Hunan Normal University, Changsha, Hunan Province, China
| | - Yong Zhang
- Medical College of Hunan Normal University, Changsha, Hunan Province, China
| | - Lesai Li
- Hunan Cancer Hospital, Changsha, Hunan Province, China
| |
Collapse
|
33
|
Allen-Gondringer A, Gau D, Dutta P, Roy P. Haplo-insufficiency of Profilin1 in vascular endothelial cells is beneficial but not sufficient to confer protection against experimentally induced atherosclerosis. Cytoskeleton (Hoboken) 2024:10.1002/cm.21859. [PMID: 38623956 PMCID: PMC11480255 DOI: 10.1002/cm.21859] [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: 10/31/2023] [Revised: 03/12/2024] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Abstract
Actin cytoskeleton plays an important role in various aspects of atherosclerosis, a key driver of ischemic heart disease. Actin-binding protein Profilin1 (Pfn1) is overexpressed in atherosclerotic plaques in human disease, and Pfn1, when partially depleted globally in all cell types, confers atheroprotection in vivo. This study investigates the impact of endothelial cell (EC)-specific partial loss of Pfn1 expression in atherosclerosis development. We utilized mice engineered for conditional heterozygous knockout of the Pfn1 gene in ECs, with atherosclerosis induced by depletion of hepatic LDL receptor by gene delivery of PCSK9 combined with high-cholesterol diet. Our studies show that partial depletion of EC Pfn1 has certain beneficial effects marked by dampening of select pro-atherogenic cytokines (CXCL10 and IL7) with concomitant reduction in cytotoxic T cell abundance but is not sufficient to reduce hyperlipidemia and confer atheroprotection in vivo. In light of these findings, we conclude that atheroprotective phenotype conferred by global Pfn1 haplo-insufficiency requires contributions of additional cell types that are relevant for atherosclerosis progression.
Collapse
Affiliation(s)
| | - David Gau
- Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | - Partha Dutta
- Bioengineering, University of Pittsburgh, Pittsburgh, PA
- Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Partha Roy
- Bioengineering, University of Pittsburgh, Pittsburgh, PA
- Pathology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
34
|
Kamisah Y, Che Hassan HH. Role of Trimetazidine in Ameliorating Endothelial Dysfunction: A Review. Pharmaceuticals (Basel) 2024; 17:464. [PMID: 38675424 PMCID: PMC11054808 DOI: 10.3390/ph17040464] [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: 03/12/2024] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Endothelial dysfunction is a hallmark of cardiovascular diseases, contributing to impaired vasodilation, altered hemodynamics, and atherosclerosis progression. Trimetazidine, traditionally used for angina pectoris, exhibits diverse therapeutic effects on endothelial dysfunction. This review aims to elucidate the mechanisms underlying trimetazidine's actions and its potential as a therapeutic agent for endothelial dysfunction and associated cardiovascular disorders. Trimetazidine enhances vasodilation and hemodynamic function by modulating endothelial nitric oxide synthase activity, nitric oxide production, and endothelin-1. It also ameliorates metabolic parameters, including reducing blood glucose, mitigating oxidative stress, and dampening inflammation. Additionally, trimetazidine exerts antiatherosclerotic effects by inhibiting plaque formation and promoting its stability. Moreover, it regulates apoptosis and angiogenesis, fostering endothelial cell survival and neovascularization. Understanding trimetazidine's multifaceted mechanisms underscores its potential as a therapeutic agent for endothelial dysfunction and associated cardiovascular disorders, warranting further investigation for clinical translation.
Collapse
Affiliation(s)
- Yusof Kamisah
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Hamat H. Che Hassan
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
35
|
Kumarapperuma H, Wang R, Little PJ, Kamato D. Mechanistic insight: Linking cardiovascular complications of inflammatory bowel disease. Trends Cardiovasc Med 2024; 34:203-211. [PMID: 36702388 DOI: 10.1016/j.tcm.2023.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 01/25/2023]
Abstract
Cardiovascular diseases (CVD) are the leading cause of mortality worldwide despite an aggressive reduction of traditional cardiovascular risk factors. Underlying inflammatory conditions such as inflammatory bowel disease (IBD) increase the risk of developing CVD. A broad understanding of the underlying pathophysiological processes between IBD and CVD is required to treat and prevent cardiovascular events in patients with IBD. This review highlights the commonality between IBD and CVD, including dysregulated immune response, genetics, environmental risk factors, altered gut microbiome, stress, endothelial dysfunction and abnormalities, to shed light on an essential area of modern medicine.
Collapse
Affiliation(s)
- Hirushi Kumarapperuma
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia; Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia
| | - Ran Wang
- Mater Research Institute, The University of Queensland, Translational Research Institute, Queensland 4102, Australia
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China
| | - Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia; Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia; School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia.
| |
Collapse
|
36
|
Niu Q, Zhao L, Wang R, Du L, Shi Q, Jia C, Li G, Jin L, Li F. Predictive value of contrast-enhanced ultrasonography and ultrasound elastography for management of BI-RADS category 4 nonpalpable breast masses. Eur J Radiol 2024; 173:111391. [PMID: 38422608 DOI: 10.1016/j.ejrad.2024.111391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
PURPOSE The objective of this study was to investigate the independent risk factors and associated predictive values of contrast-enhanced ultrasound (CEUS), shear wave elastography (SWE), and strain elastography (SE) for high-risk lesions (HRL) and malignant tumors (MT) among nonpalpable breast masses classified as BI-RADS category 4 on conventional ultrasound. METHODS This prospective study involved consecutively admitted patients with breast tumors from January 2018, aiming to explore the management of BI-RADS category 4 breast tumors using CEUS and elastography. We conducted a retrospective review of patient data, focusing on those with a history of a nonpalpable mass as the primary complaint. Pathologic findings after surgical resection served as the gold standard. The CEUS arterial-phase indices were analyzed using contrast agent arrival-time parametric imaging processing mode, while quantitative and qualitative indices were examined on ES images. Independent risk factors were identified through binary logistic regression multifactorial analysis. The predictive efficacy of different modalities was compared using a receiver operating characteristics curve. Subsequently, a nomogram for predicting the risk of HRL/MT was established based on a multifactorial logistic regression model. RESULTS A total of 146 breast masses from 146 patients were included, comprising 80 benign tumors, 12 HRLs, and 54 MTs based on the final pathology. There was no significant difference in pathologic size between the benign and HRL/MT groups [8.00(6.25,10.00) vs. 9.00(6.00,10.00), P = 0.506]. The diagnostic efficacy of US plus CEUS exceeded that of US plus SWE/SE for BI-RADS 4 nonpalpable masses, with an AUC of 0.954 compared to 0.798/0.741 (P < 0.001). Further stratified analysis revealed a more pronounced improvement for reclassification of BI-RADS 4a masses (AUC: 0.943 vs. 0.762/0.675, P < 0.001) than BI-RADS 4b (AUC:0.950 vs. 0.885/0.796, P>0.05) with the assistance of CEUS than SWE/SE. Employing downgrade CEUS strategies resulted in negative predictive values ranging from 95.2 % to 100.0 % for BI-RADS 4a and 4b masses. Conversely, using upgrade nomogram strategies, which included the independent predictive risk factors of irregular enhanced shape, poor defined enhanced margin, earlier enhanced time, increased surrounding vessels, and presence of contrast agent retention, the diagnostic performance achieved an AUC of 0.947 with good calibration. CONCLUSION After investigating the potential of CEUS and ES in improving risk assessment and diagnostic accuracy for nonpalpable BI-RADS category 4 breast masses, it is evident that CEUS has a more significant impact on enhancing classification compared to ES, particularly for BI-RADS 4a subgroup masses. This finding suggests that CEUS may offer greater benefits in improving risk assessment and diagnostic accuracy for this specific subgroup of breast masses.
Collapse
Affiliation(s)
- Qinghua Niu
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Zhao
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruitao Wang
- Department of Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lianfang Du
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiusheng Shi
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Jia
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gang Li
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lifang Jin
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Fan Li
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
37
|
Ayalew BD, Rodoshi ZN, Patel VK, Alresheq A, Babu HM, Aurangzeb RF, Aurangzeb RI, Mdivnishvili M, Rehman A, Shehryar A, Hassan A. Nuclear Cardiology in the Era of Precision Medicine: Tailoring Treatment to the Individual Patient. Cureus 2024; 16:e58960. [PMID: 38800181 PMCID: PMC11127713 DOI: 10.7759/cureus.58960] [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] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Nuclear cardiology, employing advanced imaging technologies like positron emission tomography (PET) and single photon emission computed tomography (SPECT), is instrumental in diagnosing, risk stratifying, and managing heart diseases. Concurrently, precision medicine advocates for treatments tailored to each patient's genetic, environmental, and lifestyle specificities, promising a revolution in personalized cardiovascular care. This review explores the synergy between nuclear cardiology and precision medicine, highlighting advancements, potential enhancements in patient outcomes, and the challenges and opportunities of this integration. We examined the evolution of nuclear cardiology technologies, including PET and SPECT, and their role in cardiovascular diagnostics. We also delved into the principles of precision medicine, focusing on genetic and molecular profiling, data analytics, and individualized treatment strategies. The integration of these domains aims to optimize diagnostic accuracy, therapeutic interventions, and prognostic evaluations in cardiovascular care. Advancements in molecular imaging and the application of artificial intelligence in nuclear cardiology have significantly improved the precision of diagnostics and treatment plans. The adoption of precision medicine principles in nuclear cardiology enables the customization of patient care, leveraging genetic information and biomarkers for enhanced therapeutic outcomes. However, challenges such as data integration, accessibility, cost, and the need for specialized expertise persist. The confluence of nuclear cardiology and precision medicine offers a promising pathway toward revolutionizing cardiovascular healthcare, providing more accurate, effective, and personalized patient care. Addressing existing challenges and fostering interdisciplinary collaboration is crucial for realizing the full potential of this integration in improving patient outcomes.
Collapse
Affiliation(s)
- Biruk D Ayalew
- Internal Medicine, Saint Paul's Hospital Millennium Medical College, Addis Ababa, ETH
| | | | | | - Alaa Alresheq
- Primary Care, United Nations for Relief and Works Agency, Ramallah, PSE
| | - Hisham M Babu
- Internal Medicine, Jagadguru Sri Shivarathreeshwara (JSS) Medical College and Hospital, JSS Academy of Higher Education and Research (JSSAHER), Mysore, IND
| | | | | | | | | | | | | |
Collapse
|
38
|
Stangret A, Sadowski KA, Jabłoński K, Kochman J, Opolski G, Grabowski M, Tomaniak M. Chemokine Fractalkine and Non-Obstructive Coronary Artery Disease-Is There a Link? Int J Mol Sci 2024; 25:3885. [PMID: 38612695 PMCID: PMC11012077 DOI: 10.3390/ijms25073885] [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: 02/17/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Non-obstructive coronary artery disease (NO-CAD) constitutes a heterogeneous group of conditions collectively characterized by less than 50% narrowing in at least one major coronary artery with a fractional flow reserve (FFR) of ≤0.80 observed in coronary angiography. The pathogenesis and progression of NO-CAD are still not fully understood, however, inflammatory processes, particularly atherosclerosis and microvascular dysfunction are known to play a major role in it. Chemokine fractalkine (FKN/CX3CL1) is inherently linked to these processes. FKN/CX3CL1 functions predominantly as a chemoattractant for immune cells, facilitating their transmigration through the vessel wall and inhibiting their apoptosis. Its concentrations correlate positively with major cardiovascular risk factors. Moreover, promising preliminary results have shown that FKN/CX3CL1 receptor inhibitor (KAND567) administered in the population of patients with ST-elevation myocardial infarction (STEMI) undergoing percutaneous coronary intervention (PCI), inhibits the adverse reaction of the immune system that causes hyperinflammation. Whereas the link between FKN/CX3CL1 and NO-CAD appears evident, further studies are necessary to unveil this complex relationship. In this review, we critically overview the current data on FKN/CX3CL1 in the context of NO-CAD and present the novel clinical implications of the unique structure and function of FKN/CX3CL1 as a compound which distinctively contributes to the pathomechanism of this condition.
Collapse
Affiliation(s)
- Aleksandra Stangret
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland;
| | - Karol Artur Sadowski
- 1st Department of Cardiology, Medical University of Warsaw, Banacha 1a, 01-267 Warsaw, Poland; (K.A.S.); (K.J.); (J.K.); (G.O.); (M.G.)
| | - Konrad Jabłoński
- 1st Department of Cardiology, Medical University of Warsaw, Banacha 1a, 01-267 Warsaw, Poland; (K.A.S.); (K.J.); (J.K.); (G.O.); (M.G.)
| | - Janusz Kochman
- 1st Department of Cardiology, Medical University of Warsaw, Banacha 1a, 01-267 Warsaw, Poland; (K.A.S.); (K.J.); (J.K.); (G.O.); (M.G.)
| | - Grzegorz Opolski
- 1st Department of Cardiology, Medical University of Warsaw, Banacha 1a, 01-267 Warsaw, Poland; (K.A.S.); (K.J.); (J.K.); (G.O.); (M.G.)
| | - Marcin Grabowski
- 1st Department of Cardiology, Medical University of Warsaw, Banacha 1a, 01-267 Warsaw, Poland; (K.A.S.); (K.J.); (J.K.); (G.O.); (M.G.)
| | - Mariusz Tomaniak
- 1st Department of Cardiology, Medical University of Warsaw, Banacha 1a, 01-267 Warsaw, Poland; (K.A.S.); (K.J.); (J.K.); (G.O.); (M.G.)
| |
Collapse
|
39
|
Bagheri B, Khatibiyan Feyzabadi Z, Nouri A, Azadfallah A, Mahdizade Ari M, Hemmati M, Darban M, Alavi Toosi P, Banihashemian SZ. Atherosclerosis and Toll-Like Receptor4 (TLR4), Lectin-Like Oxidized Low-Density Lipoprotein-1 (LOX-1), and Proprotein Convertase Subtilisin/Kexin Type9 (PCSK9). Mediators Inflamm 2024; 2024:5830491. [PMID: 38445291 PMCID: PMC10914434 DOI: 10.1155/2024/5830491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/31/2024] [Accepted: 02/16/2024] [Indexed: 03/07/2024] Open
Abstract
Atherosclerosis is a leading cause of death in the world. A significant body of evidence suggests that inflammation and various players are implicated and have pivotal roles in the formation of atherosclerotic plaques. Toll-like receptor 4 (TLR4) is linked with different stages of atherosclerosis. This receptor is highly expressed in the endothelial cells (ECs) and atherosclerotic plaques. TLR4 activation can lead to the production of inflammatory cytokines and related responses. Lectin-like oxidized low-density lipoprotein-1 (LOX-1), an integral membrane glycoprotein with widespread expression on the ECs, is involved in atherosclerosis and has some common pathways with TLR4 in atherosclerotic lesions. In addition, proprotein convertase subtilisin/kexin type9 (PCSK9), which is a regulatory enzyme with different roles in cholesterol uptake, is implicated in atherosclerosis. At present, TLR4, PCSK9, and LOX-1 are increasingly acknowledged as key players in the pathogenesis of atherosclerotic cardiovascular diseases. Herein, we presented the current evidence on the structure, functions, and roles of TLR4, PCSK9, and LOX-1 in atherosclerosis.
Collapse
Affiliation(s)
- Bahador Bagheri
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Ahmad Nouri
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Azadfallah
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mahyar Mahdizade Ari
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maral Hemmati
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Mahboubeh Darban
- Department of Internal Medicine, Kowsar Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Parisa Alavi Toosi
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | |
Collapse
|
40
|
Sharma P, Roy A, Dhamija RK, Bhushan S, Baswal K, Kulandaisamy R, Yadav S, Kumar S, Inampudi KK. A comprehensive proteomic profiling of urinary exosomes and the identification of early non-invasive biomarker in patients with coronary artery disease. J Proteomics 2024; 293:105059. [PMID: 38151158 DOI: 10.1016/j.jprot.2023.105059] [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: 10/05/2023] [Revised: 12/04/2023] [Accepted: 12/14/2023] [Indexed: 12/29/2023]
Abstract
Urinary small extracellular vesicles or exosomes (uEVs) source could be an emerging trove of biomarkers in coronary artery disease (CAD). It is a chronic inflammatory disease having a long asymptomatic phase of fatty-fibrous development in arteries leading to angina, myocardial infarction, and death. Our study was aimed at identifying differential protein expression profiling of uEVs in CAD. We collected urine samples of CAD patients (n = 41) age 18-65 years and gender matched healthy controls (n = 41). We isolated uEVs using differential ultracentrifugation. Further, uEV samples were characterized by western blotting exosome markers (Flotillin, TSG, CD63, and CD9), nano tracking analysis, and transmission and scanning electron microscopy. A total of 508 proteins were identified by iTRAQ-based mass spectrometry. We observed protein expression levels of AZGP1, SEMG1/2, ORM1, IGL, SERPINA5, HSPG2, prosaposin, gelsolin, and CD59 were upregulated, and UMOD, KNG1, AMBP, prothrombin, and TF were downregulated. Protein-protein interactions, gene ontology and pathway analysis were performed to functionally annotate identified uEVs proteins. A novel uEVs differential protein signature is shown. On validating UMOD protein by ELISA in two clinically different CAD, stable-CAD patients had lower levels than healthy controls whereas recent myocardial infarction patients had lowest. Our findings suggest UMOD importance as early diagnostic biomarker. SIGNIFICANCE: Coronary artery disease is a chronic inflammatory disease caused by gradual deposition of cholesterol and fat along with other proteins to develop plaque inside arteries. This further leads to blockage of artery, heart attack and death. There are no identifiable early biomarkers to diagnose this. For the first time, we have identified the differentially expressed proteins isolated from non-invasive uEV of CAD patients compared to healthy controls by using MS Orbitrap and iTRAQ labelling of peptides. We have identified decreased levels of UMOD protein in CAD. These findings have been confirmed by ELISA. Furthermore, the levels of UMOD were observed as more highly decreased in recent myocardial infarction CAD patients, indicating the importance of this protein as an early diagnostic biomarker. Conclusively, our study represents a non-invasive urinary EVs trove of differentially expressed proteins in CAD. This will form a groundwork for understanding the pathophysiology of CAD and will help in future translational research utilizing uEVs.
Collapse
Affiliation(s)
- Pratibha Sharma
- Human Behaviour Department, Institute of Human Behaviour and Allied Sciences, Dilshad Garden, New Delhi, India; Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| | - Ambuj Roy
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Rajinder K Dhamija
- Human Behaviour Department, Institute of Human Behaviour and Allied Sciences, Dilshad Garden, New Delhi, India
| | - Sudha Bhushan
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Kamal Baswal
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | | | - Satyavir Yadav
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India; Department of Health Education and Technology, Luleå University of Technology, Sweden
| | | |
Collapse
|
41
|
Nègre-Salvayre A, Salvayre R. Reactive Carbonyl Species and Protein Lipoxidation in Atherogenesis. Antioxidants (Basel) 2024; 13:232. [PMID: 38397830 PMCID: PMC10886358 DOI: 10.3390/antiox13020232] [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: 12/29/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Atherosclerosis is a multifactorial disease of medium and large arteries, characterized by the presence of lipid-rich plaques lining the intima over time. It is the main cause of cardiovascular diseases and death worldwide. Redox imbalance and lipid peroxidation could play key roles in atherosclerosis by promoting a bundle of responses, including endothelial activation, inflammation, and foam cell formation. The oxidation of polyunsaturated fatty acids generates various lipid oxidation products such as reactive carbonyl species (RCS), including 4-hydroxy alkenals, malondialdehyde, and acrolein. RCS covalently bind to nucleophilic groups of nucleic acids, phospholipids, and proteins, modifying their structure and activity and leading to their progressive dysfunction. Protein lipoxidation is the non-enzymatic post-translational modification of proteins by RCS. Low-density lipoprotein (LDL) oxidation and apolipoprotein B (apoB) modification by RCS play a major role in foam cell formation. Moreover, oxidized LDLs are a source of RCS, which form adducts on a huge number of proteins, depending on oxidative stress intensity, the nature of targets, and the availability of detoxifying systems. Many systems are affected by lipoxidation, including extracellular matrix components, membranes, cytoplasmic and cytoskeletal proteins, transcription factors, and other components. The mechanisms involved in lipoxidation-induced vascular dysfunction are not fully elucidated. In this review, we focus on protein lipoxidation during atherogenesis.
Collapse
Affiliation(s)
- Anne Nègre-Salvayre
- Inserm Unité Mixte de Recherche (UMR), 1297 Toulouse, Centre Hospitalier Universitaire (CHU) Rangueil—BP 84225, 31432 Toulouse CEDEX 4, France;
- Faculty of Medicine, University of Toulouse, 31432 Toulouse, France
| | - Robert Salvayre
- Inserm Unité Mixte de Recherche (UMR), 1297 Toulouse, Centre Hospitalier Universitaire (CHU) Rangueil—BP 84225, 31432 Toulouse CEDEX 4, France;
- Faculty of Medicine, University of Toulouse, 31432 Toulouse, France
| |
Collapse
|
42
|
Mosleh MM, Sohn MJ, Kim HS. Endothelial marker profiles in cerebral radiation-induced vasculopathy: A comparative immunohistochemical analysis. Medicine (Baltimore) 2024; 103:e37130. [PMID: 38306519 PMCID: PMC10843420 DOI: 10.1097/md.0000000000037130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/10/2024] [Indexed: 02/04/2024] Open
Abstract
Radiation therapy results in radiation-induced vasculopathy, characterized by alterations in the vascular architecture stemming from radiation exposure. The exact molecular pathways and associated pathologies of this condition have yet to be comprehensively understood. This study aimed to identify specific markers' roles in cerebral vascular endothelial injury pathogenesis after radiosurgery and explore their unique expression patterns in diverse pathologies post-stereotactic radiosurgery. A retrospective cohort study was conducted to assess the expression profiles of endothelial markers via immunohistochemical analysis in 25 adult patients (13 males and 12 females) who had undergone neurosurgical resection for various central nervous system pathologies following stereotactic radiosurgery or radiotherapy from 2001 to 2015. Our findings revealed strong immunohistochemical expression of ICAM-1 and E-selectin across various disease states, while MMP-9, PAI-1, and eNOS exhibited moderate expression levels. In contrast, VCAM-1 and P-Selectin had the weakest expression across all groups. Notably, while individual markers showed significant variations in expression levels when comparing different diseases (P < .001), no substantial differences were found in the overall immunohistochemical expression patterns across the 5 distinct pathologies studied (P = .407, via 2-way ANOVA). Despite the varied long-term effects of radiotherapy on the vascular endothelium, a common thread of inflammation runs through the pathology of these conditions. The distinct patterns of marker expression identified in our study suggest that different markers play unique roles in the development of radiation-induced vasculopathy. These findings offer insights that could lead to the development of novel preventive strategies and treatments.
Collapse
Affiliation(s)
- Mohammad Mohsen Mosleh
- Department of Biomedical Science, Graduate School of Medicine, Inje University, Busanjin-gu, Busan, Korea
| | - Moon-Jun Sohn
- Department of Biomedical Science, Graduate School of Medicine, Inje University, Busanjin-gu, Busan, Korea
- Department of Neurosurgery and Neuroscience & Radiosurgery Hybrid Research Center, Inje University Ilsan Paik Hospital, College of Medicine, Ilsanseo-gu, Goyang City, Gyeonggi-do, Korea
| | - Han Seong Kim
- Department of Pathology, Inje University Ilsan Paik Hospital, Inje University Ilsan Paik Hospital, College of Medicine, Ilsanseo-gu, Goyang City, Gyeonggi-do, Korea
| |
Collapse
|
43
|
Gofir A, Satriotomo I, Syamsah YCBN, Rochmah MA, Setyawan TR, Mianoki A, Silalahi RANA, Nugroho DB. Degree of COVID-19 severity and mortality in stroke: correlation of clinical and laboratory parameters. BMC Neurosci 2024; 25:4. [PMID: 38216918 PMCID: PMC10790265 DOI: 10.1186/s12868-023-00837-w] [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: 10/12/2023] [Accepted: 12/07/2023] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Stroke is one of the neurological manifestations of COVID-19, leading to a significant risk of morbidity and mortality. Clinical manifestations and laboratory parameters were investigated to determine mortality predictors in this case. METHOD The case control study was conducted at Dr. Sardjito General Hospital,Yogyakarta, Indonesia, with data collected between July 2020 and August 2021. All recorded clinical and laboratory data from acute stroke patients with confirmed COVID-19 were collected. Baseline characteristics, bivariate, and multivariate analyses were assessed to determine significant predictors for mortality. RESULT This study involved 72 subjects with COVID-19 and stroke. The majority experienced ischemic stroke, with hypertension as the most prevalent comorbidity. Notably, 45.8% of subjects (p < 0.05) loss of consciousness and 72.2% of exhibited motor deficits (p < 0.05). Severe degree of COVID-19 was observed in 52.8% of patients, with respiratory distress and death rates of 56.9% and 58.3%. Comparison of surviving and deceased groups highlighted significant differences in various clinical and laboratory characteristics differences. Hazard ratio (HR) analysis identified loss of consciousness (HR = 2.68; p = 0.01), motor deficit (HR = 2.34; p = 0.03), respiratory distress (HR = 81.51; p < 0.001), and monocyte count (HR:1.002; p = 0.04) as significant predictors of mortality. CONCLUSION Mortality in COVID-19 patients with stroke was significantly associated with loss of consciousness, motor deficit, respiratory distress, and raised monocyte count. The risk of mortality is heightened when multiple factors coexist.
Collapse
Affiliation(s)
- Abdul Gofir
- Department of Neurology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada/ Dr. Sardjito General Hospital, Jalan Farmako Sekip Utara, Sleman, Mlati, Yogyakarta, 55281, Indonesia.
| | | | - Yossy Catarina Budi Nur Syamsah
- Department of Neurology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada/ Dr. Sardjito General Hospital, Jalan Farmako Sekip Utara, Sleman, Mlati, Yogyakarta, 55281, Indonesia
| | - Mawaddah Ar Rochmah
- Department of Neurology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada/ Dr. Sardjito General Hospital, Jalan Farmako Sekip Utara, Sleman, Mlati, Yogyakarta, 55281, Indonesia
| | - Tommy Rachmat Setyawan
- Department of Neurology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada/ Dr. Sardjito General Hospital, Jalan Farmako Sekip Utara, Sleman, Mlati, Yogyakarta, 55281, Indonesia
| | - Adika Mianoki
- Department of Neurology, Dr. Soeradji Tirtonegoro General Hospital, Klaten, Central Java, Indonesia
| | - Raymond Aris Nimrod Alvonsius Silalahi
- Department of Neurology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada/ Dr. Sardjito General Hospital, Jalan Farmako Sekip Utara, Sleman, Mlati, Yogyakarta, 55281, Indonesia
| | - Dhite Bayu Nugroho
- Department of Internal Medicine, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
44
|
Huang H, Sun Z, Xu J, Wang L, Zhao J, Li J, Zhang S, Yuan F, Liu M, Fang Z. Yang-Xin-Shu-Mai granule alleviates atherosclerosis by regulating macrophage polarization via the TLR9/MyD88/NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116868. [PMID: 37454749 DOI: 10.1016/j.jep.2023.116868] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/04/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Previous studies have found that Yang-Xin-Shu-Mai granule (YXSMG) has certain advantages in the treatment of stable coronary heart disease. However, YXSMG can inhibit the progression of atherosclerotic plaque and stabilize vulnerable plaque needs to be further explored and studied. This research, mass spectrometry analysis, network pharmacology, in vivo and in vitro experimental studies were conducted to explore the mechanism of YXSMG on atherosclerosis. AIM OF THE STUDY To decipher the mechanism of atherosclerotic plaque, stabilization for YXSMG by analysis of its active ingredients and biological network and activity in whole animal and at cellular and molecular levels. METHODS The active components of YXSMG were determined using high performance liquid chromatography-mass spectrometry/mass spectrometry (HPLC-MS/MS) analysis. The 'Disease-Compound-Target-Pathway' network diagram was constructed using network pharmacology, and the stability of binding between core targets and core compounds was analyzed with molecular docking. After intervention with YXSMG, the pathology of aortic plaque, inflammation in the surrounding tissue, expression of TLR9/MyD88/NF-κB pathway protein in plaque and M1/M2 polarization of plaque macrophages were evaluated in vivo in apolipoprotein E-deficient (ApoE-/-) mice fed with high-fat diet. To verify whether it suppressed inflammation by inhibiting Toll-like receptor 9 (TLR9) reprogramming of macrophage polarization, we used RAW264.7 macrophages treated with specific TLR9 agonist (ODN1826) and inhibitor (ODN2088). RESULTS Five active compounds were identified in YXSMG: catechin, formononetin, tanshinone IIA, cryptotanshinone and glycitein. Network pharmacology studies revealed TLR9 as one of the core targets of YXSMG intervention in atherosclerosis. Computer simulation of molecular docking showed that TLR9 could interact with the core compound to form a stable complex. In vivo experiments confirmed that YXSMG could significantly inhibit atherosclerotic plaque, reduce levels of blood lipids and inflammatory factors, downregulate TLR9/MyD88/NF-κB pathway protein and inhibit aortic sinus macrophages polarization to M1, but promote their polarization to M2 to inhibit inflammation. In vitro experiments revealed that YXSMG could downregulate expression of TLR9 gene and protein in ODN1826-activated RAW264.7 macrophages. ODN2088 had a synergistic effect with YXSMG on the TLR9/MyD88/NF-κB signaling pathway, and reprogrammed macrophages polarization from M1 to M2 by inhibiting TLR9, thus reducing immuno-inflammatory response. CONCLUSION YXSMG can reduce the level of blood lipid and improve the size of atherosclerotic plaque and inflammatory infiltration in ApoE-/- mice fed with high fat. It is concluded that YXSMG can improve the mechanism of atherosclerotic plaque by inhibiting TLR9/MyD88/NF-κB pathway reprogramming macrophage M1/M2 polarization and reducing arterial inflammation.
Collapse
Affiliation(s)
- Hong Huang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China.
| | - Zeqi Sun
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China.
| | - Junyao Xu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China
| | - Linjie Wang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China
| | - Jing Zhao
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China
| | - Jie Li
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China
| | - Siqi Zhang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China; Institute of Hypertension, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China
| | - Fang Yuan
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China
| | - Ming Liu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China; Institute of Hypertension, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China.
| | - Zhuyuan Fang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China; Institute of Hypertension, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China.
| |
Collapse
|
45
|
Di Muzio C, Di Cola I, Shariat Panahi A, Ursini F, Iagnocco A, Giacomelli R, Cipriani P, Ruscitti P. The effects of suppressing inflammation by tofacitinib may simultaneously improve glycaemic parameters and inflammatory markers in rheumatoid arthritis patients with comorbid type 2 diabetes: a proof-of-concept, open, prospective, clinical study. Arthritis Res Ther 2024; 26:14. [PMID: 38178250 PMCID: PMC10765862 DOI: 10.1186/s13075-023-03249-7] [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: 11/04/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND A consistent connection has been increasingly reported between rheumatoid arthritis (RA), insulin resistance (IR), and type 2 diabetes (T2D). The β-cell apoptosis induced by pro-inflammatory cytokines, which could be exaggerated in the context of RA, is associated with increased expression pro-apoptotic proteins, which is dependent on JAnus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) activation. On these bases, we aimed to evaluate if the administration of tofacitinib, a potent and selective JAK inhibitor, could simultaneously improve glycaemic parameters and inflammatory markers in patients with RA and comorbid T2D. METHODS The primary endpoint was the change in the 1998-updated homeostatic model assessment of IR (HOMA2-IR) after 6 months of treatment with tofacitinib in RA patients with T2D. Consecutive RA patients with T2D diagnosis were included in this proof-of-concept, open, prospective, clinical study, which was planned before the recent emergence of safety signals about tofacitinib. Additional endpoints were also assessed regarding RA disease activity and metabolic parameters. RESULTS Forty consecutive RA patients with T2D were included (female sex 68.9%, mean age of 63.4 ± 9.9 years). During 6-month follow-up, a progressive reduction of HOMA2-IR was observed in RA patients with T2D treated with tofacitinib. Specifically, a significant effect of tofacitinib was shown on the overall reduction of HOMA2-IR (β = - 1.1, p = 0.019, 95%CI - 1.5 to - 0.76). Also, HOMA2-β enhanced in these patients highlighting an improvement of insulin sensitivity. Furthermore, although a longer follow-up is required, a trend in glycated haemoglobin reduction was also recorded. The administration of tofacitinib induced an improvement in RA disease activity, and a significant reduction of DAS28-CRP and SDAI was observed; 76.8% of patients achieved a good clinical response. In this study, no major adverse events (AEs) were retrieved without the identification of new safety signals. Specifically, no life-threatening AEs and cardiovascular and/or thromboembolic events were recorded. CONCLUSIONS The administration of tofacitinib in RA with T2D led to a simultaneous improvement of IR and inflammatory disease activity, inducing a "bidirectional" benefit in these patients. However, further specific designed and powered studies are warranted to entirely evaluate the metabolic effects of tofacitinib in RA patients with T2D.
Collapse
Affiliation(s)
- Claudia Di Muzio
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, PO box 67100, L'Aquila, Italy
| | - Ilenia Di Cola
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, PO box 67100, L'Aquila, Italy
| | - Azadeh Shariat Panahi
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, PO box 67100, L'Aquila, Italy
| | - Francesco Ursini
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Annamaria Iagnocco
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Roberto Giacomelli
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128, Rome, Italy
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Paola Cipriani
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, PO box 67100, L'Aquila, Italy
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, PO box 67100, L'Aquila, Italy.
| |
Collapse
|
46
|
Pathak A, Pal AK, Roy S, Nandave M, Jain K. Role of Angiogenesis and Its Biomarkers in Development of Targeted Tumor Therapies. Stem Cells Int 2024; 2024:9077926. [PMID: 38213742 PMCID: PMC10783989 DOI: 10.1155/2024/9077926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 01/13/2024] Open
Abstract
Angiogenesis plays a significant role in the human body, from wound healing to tumor progression. "Angiogenic switch" indicates a time-restricted event where the imbalance between pro- and antiangiogenic factors results in the transition from prevascular hyperplasia to outgrowing vascularized tumor, which eventually leads to the malignant cancer progression. In the last decade, molecular players, i.e., angiogenic biomarkers and underlying molecular pathways involved in tumorigenesis, have been intensely investigated. Disrupting the initiation and halting the progression of angiogenesis by targeting these biomarkers and molecular pathways has been considered as a potential treatment approach for tumor angiogenesis. This review discusses the currently known biomarkers and available antiangiogenic therapies in cancer, i.e., monoclonal antibodies, aptamers, small molecular inhibitors, miRNAs, siRNAs, angiostatin, endostatin, and melatonin analogues, either approved by the U.S. Food and Drug Administration or currently under clinical and preclinical investigations.
Collapse
Affiliation(s)
- Anchal Pathak
- Drug Delivery and Nanomedicine Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, India
| | - Ajay Kumar Pal
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Mukesh Nandave
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India
| | - Keerti Jain
- Drug Delivery and Nanomedicine Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, India
| |
Collapse
|
47
|
Zou X, Li Y, Yang J, Miao J, Li Y, Ling W. Contrast-enhanced ultrasound reveals free-floating thrombus in carotid artery: The cause of stroke is surprisingly plaque rupture. Clin Hemorheol Microcirc 2024; 87:129-136. [PMID: 38277285 DOI: 10.3233/ch-232037] [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: 01/28/2024]
Abstract
BACKGROUND Acute stroke poses a serious threat to people's health. The occurrence of a thrombus following the rupture of vulnerable plaques in the carotid artery is a significant contributor to the development of stroke. In previous case reports, it has been challenging to visualize tiny ulcerations within carotid artery plaques using computed tomography angiography (CTA) and digital subtraction angiography (DSA), even when the rupture of the plaque leads to the formation of a free-floating thrombus (FFT). However, in this particular case, contrast-enhanced ultrasound (CEUS) was able to overcome this limitation and provide a more precise assessment, confirming that the FFT formation was indeed a result of plaque rupture rather than any other potential causes. Cases that utilize CEUS to visualize the formation of ulcers and FFT resulting from plaque rupture are even more rare. As such, we present this case to shed light on this infrequent phenomenon. CASE SUMMARY In this case study, we present a 65-year-old male patient who was admitted to the hospital due to headache and abnormal mental behavior for one day. During the routine cervical artery ultrasound examination upon admission, we detected the presence of plaque in the right internal carotid artery of the patient, resulting in luminal stenosis. Additionally, we observed suspected hypoechoic material at the distal end of the plaque. After undergoing CEUS examination, it was definitively determined that an ulcer had formed and a FFT had developed due to the rupture of carotid artery plaque. Subsequent CTA and DSA examinations further confirmed the presence of the FFT. The magnetic resonance imaging (MRI) reveals an acute lacunar infarction in the head of the right caput nuclei caudate, which strengthens the potential link between the patient's neurological and psychiatric symptoms observed during admission. The patient received prompt antiplatelet therapy and underwent cervical artery stenting surgery with the assistance of a distal embolic protection device. Following the procedure, the patient was discharged on the fourth day and experienced a complete recovery. CONCLUSION CEUS is a valuable tool for visualizing FFT resulting from the rupture of vulnerable plaques in the carotid artery.
Collapse
Affiliation(s)
- Xiuli Zou
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Medical Ultrasound, Zigong Fourth People's Hospital, Zigong, Sichuan, China
| | - Ying Li
- Department of Medical Ultrasound, Zigong Fourth People's Hospital, Zigong, Sichuan, China
| | - Jilan Yang
- Department of Medical Ultrasound, Zigong Fourth People's Hospital, Zigong, Sichuan, China
| | - Juan Miao
- Department of Medical Ultrasound, Zigong Fourth People's Hospital, Zigong, Sichuan, China
| | - Yuan Li
- Department of Medical Ultrasound, Zigong Fourth People's Hospital, Zigong, Sichuan, China
| | - Wenwu Ling
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
48
|
Qiao Q, Wang B, Xu M, Qi Z. Contrast-enhanced ultrasound evaluation of plaque vulnerability and the relationship between peripheral blood leukocytes. Clin Hemorheol Microcirc 2024; 87:187-197. [PMID: 38427471 DOI: 10.3233/ch-232034] [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: 03/03/2024]
Abstract
OBJECTIVE To evaluate plaque vulnerability by carotid contrast-enhanced ultrasound (CEUS) and to analyze the correlation between plaque vulnerability and peripheral blood leukocyte classification. MATERIALS AND METHODS 135 patients with carotid plaque were examined by contrast-enhanced ultrasound. Plaque vulnerability was assessed by semiquantitative visual classification. Baseline clinical data and peripheral leukocyte classification were collected. Ordered logistic regression was used to analyze the correlation between plaque neovascularization grade and peripheral leukocyte classification count. RESULTS There were significant differences in leukocyte, monocyte, neutrophil, mean platelet volume, lymphocyte, and neutrophil counts between different neovascular plaque grades and peripheral blood (P < 0.05). Correlation analysis showed that leukocyte, monocyte, and neutrophil counts were significantly positively correlated. CONCLUSION The increase in plaque neovascularization is associated with an increase in circulating leukocytes, monocytes, and neutrophils. Therefore, CEUS combined with peripheral blood leukocytes may serve as an early warning of plaque vulnerability and provide a theoretical basis for clinical treatment.
Collapse
Affiliation(s)
- Qi Qiao
- Hebei Medical University, Shijiazhuang, China
- Department of Ultrasound, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Bingshuang Wang
- The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Minzhe Xu
- Hebei Medical University, Shijiazhuang, China
- Department of Ultrasound, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Zhengqin Qi
- Department of Ultrasound, First Hospital of Qinhuangdao, Qinhuangdao, China
| |
Collapse
|
49
|
Wang W, Li H, Shi Y, Zhou J, Khan GJ, Zhu J, Liu F, Duan H, Li L, Zhai K. Targeted intervention of natural medicinal active ingredients and traditional Chinese medicine on epigenetic modification: Possible strategies for prevention and treatment of atherosclerosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155139. [PMID: 37863003 DOI: 10.1016/j.phymed.2023.155139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/26/2023] [Accepted: 10/05/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Atherosclerosis is a deadly consequence of cardiovascular disease and has very high mortality rate worldwide. The epigenetic modifications can regulate the pervasiveness and progression of atherosclerosis through its involvement in regulation of inflammation, oxidative stress, lipid metabolism and several other factors. Specific non-coding RNAs, DNA methylation, and histone modifications are key regulatory factors of atherosclerosis. Natural products from traditional Chinese medicine have shown promising therapeutic potential against atherosclerosis by means of regulating the expression of specific genes, stabilizing arterial plaques and protecting vascular endothelial cells. OBJECTIVE Our study is focusing to explore the pathophysiology and probability of traditional Chinese medicine and natural medicinal active ingredients to treat atherosclerosis. METHODS Comprehensive literature review was conducted using PubMed, Web of Science, Google Scholar and China National Knowledge Infrastructure with a core focus on natural medicinal active ingredients and traditional Chinese medicine prying in epigenetic modification related to atherosclerosis. RESULTS Accumulated evidence demonstrated that natural medicinal active ingredients and traditional Chinese medicine have been widely studied as substances that can regulate epigenetic modification. They can participate in the occurrence and development of atherosclerosis through inflammation, oxidative stress, lipid metabolism, cell proliferation and migration, macrophage polarization and autophagy respectively. CONCLUSION The function of natural medicinal active ingredients and traditional Chinese medicine in regulating epigenetic modification may provide a new potential strategy for the prevention and treatment of atherosclerosis. However, more extensive research is essential to determine the potential of these natural medicinal active ingredients to treat atherosclerosis because of least clinical data.
Collapse
Affiliation(s)
- Wei Wang
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China; College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China
| | - Han Li
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China; College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China
| | - Ying Shi
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China; College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China
| | - Jing Zhou
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China; College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China
| | - Ghulam Jilany Khan
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, University of Central Punjab, Lahore 54000, Pakistan
| | - Juan Zhu
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China; Faculty of Pharmacy, Bengbu Medical College, Bengbu, Anhui 233030, China
| | - Fawang Liu
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, Anhui 230012, China
| | - Hong Duan
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China; College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China.
| | - Lili Li
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou 234000, China.
| | - Kefeng Zhai
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China; College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China; Faculty of Pharmacy, Bengbu Medical College, Bengbu, Anhui 233030, China.
| |
Collapse
|
50
|
Zhang Y, Jiang C, Meng N. Targeting Ferroptosis: A Novel Strategy for the Treatment of Atherosclerosis. Mini Rev Med Chem 2024; 24:1262-1276. [PMID: 38284727 DOI: 10.2174/0113895575273164231130070920] [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: 08/09/2023] [Revised: 10/04/2023] [Accepted: 10/11/2023] [Indexed: 01/30/2024]
Abstract
Since ferroptosis was reported in 2012, its application prospects in various diseases have been widely considered, initially as a treatment direction for tumors. Recent studies have shown that ferroptosis is closely related to the occurrence and development of atherosclerosis. The primary mechanism is to affect the occurrence and development of atherosclerosis through intracellular iron homeostasis, ROS and lipid peroxide production and metabolism, and a variety of intracellular signaling pathways. Inhibition of ferroptosis is effective in inhibiting the development of atherosclerosis, and it can bring a new direction for treating atherosclerosis. In this review, we discuss the mechanism of ferroptosis and focus on the relationship between ferroptosis and atherosclerosis, summarize the different types of ferroptosis inhibitors that have been widely studied, and discuss some issues worthy of attention in the treatment of atherosclerosis by targeting ferroptosis.
Collapse
Affiliation(s)
- Yifan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Chengshi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Ning Meng
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| |
Collapse
|