1
|
Wu Z, Liu R, Chen J, Cai X, Yi J, Wang J, Wang D, Hu M. An enzymatic cleavage-triggered minimally invasive nanosensor for urine-based detection of early atherosclerosis. SCIENCE ADVANCES 2025; 11:eadu7614. [PMID: 40085714 PMCID: PMC11908493 DOI: 10.1126/sciadv.adu7614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
Timely detection of early atherosclerosis (AS) is crucial for improving cardiovascular outcomes, creating a growing demand for diagnostic tools that are simple, sensitive, and cost-effective. Here, we introduce a synthetic nanosensor for early AS detection that leverages the fluorescence and renal clearance properties of carbon quantum dots (CQDs). This nanosensor, designed to respond to the proteolytic activity of AS-associated dysregulated enzymes, entails CQDs as signal reporters to convert AS-associated proteolytic activity to fluorometric readings enabling a sensitive and cost-effective urine-based assay for early AS detection. Our findings demonstrated that the nanosensor provided distinct signals in atherosclerotic versus healthy mice at early AS stages, indicating its diagnostic potential. Moreover, toxicity tests showed no notable adverse effects, supporting its safety for diagnostic applications. This minimally invasive diagnostic approach could facilitate personalized therapy design and continuous efficacy assessment. It is expected that such a modular nanosensor platform can be integrated with simple urine tests to offer cost-effective detection of various diseases.
Collapse
Affiliation(s)
- Zhina Wu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China
| | - Rui Liu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Jianai Chen
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Xueying Cai
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | | | - Jiasi Wang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Min Hu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China
| |
Collapse
|
2
|
Khowdiary MM, Al-Kuraishy HM, Al-Gareeb AI, Albuhadily AK, Elhenawy AA, Rashwan EK, Alexiou A, Papadakis M, Fetoh MEAE, Batiha GES. The Peripheral Amyloid-β Nexus: Connecting Alzheimer's Disease with Atherosclerosis through Shared Pathophysiological Mechanisms. Neuromolecular Med 2025; 27:20. [PMID: 40032716 DOI: 10.1007/s12017-025-08836-2] [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: 11/13/2024] [Accepted: 02/06/2025] [Indexed: 03/05/2025]
Abstract
Alzheimer's disease (AD) and atherosclerosis (AS) are two chronic diseases with seemingly distinct pathologies. However, emerging research points to a bidirectional relationship driven by common mechanisms, such as inflammation, oxidative stress, and dysregulation of Amyloid-Beta (Aβ). This review focuses on the role of Aβ as a critical molecular link between AD and AS, emphasizing its contribution to neuronal impairment and vascular damage. Specifically, peripheral Aβ produced in the pancreas and skeletal muscle tissues exacerbates AS by promoting endothelial dysfunction and insulin resistance (IR). Furthermore, AS accelerates AD progression by impairing cerebral blood flow and inducing chronic hypoxia, causing Aβ accumulation. This review critically evaluates recent findings, highlighting inconsistencies in clinical studies and suggesting future research directions. Understanding the bidirectional influence of AD and AS could pave the way for novel therapeutic approaches targeting shared molecular pathways, particularly emphasizing Aβ clearance and inflammation.
Collapse
Affiliation(s)
- Manal M Khowdiary
- Department of Chemistry, Faculty of Applied Science, Lieth Collage, Umm Al-Qura University, 24382, Makkah, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir Ibn Hayyan Medical University, Al-Ameer Qu./Najaf-Iraq, PO. Box13, Kufa, Iraq
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ahmed A Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
- Chemistry Department, Faculty of Science, AlBaha University, 65731, Al Bahah, Saudi Arabia
| | - Eman K Rashwan
- Department of Physiology, College of Medicine, Jouf University, Akaka, Saudi Arabia
| | - Athanasios Alexiou
- Department of Research & Development, Funogen, 11741, Athens, Attiki, Greece
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
| | - Marios Papadakis
- University Hospital, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohammed E Abo-El Fetoh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Badr City, 11829, Cairo, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
3
|
Qin YS, Yi J, Chen YJ, Zhang W, Tang SF. Recent Advances in Micro/Nanomotor for the Therapy and Diagnosis of Atherosclerosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11443-11468. [PMID: 39648908 DOI: 10.1021/acsami.4c15165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Atherosclerotic cardiovascular disease poses a significant global public health threat with a high incidence that can result in severe mortality and disability. The lack of targeted effects from traditional therapeutic drugs on atherosclerosis may cause damage to other organs and tissues, necessitating the need for a more focused approach to address this dilemma. Micro/nanomotors are self-propelled micro/nanoscale devices capable of converting external energy into autonomous movement, which offers advantages in enhancing penetration depth and retention while increasing contact area with abnormal sites, such as atherosclerotic plaque, inflammation, and thrombosis, within blood vessel walls. Recent studies have demonstrated the crucial role micro/nanomotors play in treating atherosclerotic cardiovascular disease. Hence, this review highlights the recent progress of micro/nanomotor technology in atherosclerotic cardiovascular disease, including the effective promotion of micro/nanomotors in the circulatory system, overcoming hemorheological barriers, targeting the atherosclerotic plaque microenvironment, and targeting intracellular drug delivery, to facilitate atherosclerotic plaque localization and therapy. Furthermore, we also describe the potential application of micro/nanomotors in the imaging of vulnerable plaque. Finally, we discuss key challenges and prospects for treating atherosclerotic cardiovascular disease while emphasizing the importance of designing individualized management strategies specific to its causes and microenvironmental factors.
Collapse
Affiliation(s)
- Yu-Sheng Qin
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| | - Juan Yi
- Department of Laboratory Medicine, Liuzhou Traditional Chinese Medical Hospital, The Third Affiliated Hospital of Guangxi University of Chinese Medicine, Liuzhou 545006, China
| | - Yan-Jun Chen
- Department of Pathology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wei Zhang
- Department of Radiology, Liuzhou People's Hospital, Liuzhou 545006, China
| | - Shi-Fu Tang
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| |
Collapse
|
4
|
Jiang X, Huang H. The therapeutic potential of apigenin against atherosclerosis. Heliyon 2025; 11:e41272. [PMID: 39811295 PMCID: PMC11732486 DOI: 10.1016/j.heliyon.2024.e41272] [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: 09/10/2024] [Revised: 12/02/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
Apigenin is a natural flavonoid abundantly found in fruits, vegetables, and medicinal plants. It possesses protective effects against cancer, metabolic syndrome, dyslipidemia, etc. Atherosclerosis, a chronic immune-mediated inflammatory disease, is the underlying cause of coronary heart disease, stroke, and myocardial infarction. Numerous in vivo and in vitro studies have shown a protective effect of apigenin against atherosclerosis, attributed to its antioxidant and anti-inflammatory properties, as well as its antihypertensive effect and regulation of lipid metabolism. This study aimed to review the effects and mechanisms of apigenin against atherosclerosis for the first time. Apigenin displays encouraging results, and this review confirms the potential value of apigenin as a candidate medication for atherosclerosis.
Collapse
Affiliation(s)
- Xueqiang Jiang
- Sinopharm Dongfeng General Hospital, Hubei Clinical Research Center of Hypertension, Hubei University of Medicine, Shiyan, 442008, China
| | - Huimin Huang
- Sinopharm Dongfeng General Hospital, Hubei Clinical Research Center of Hypertension, Hubei University of Medicine, Shiyan, 442008, China
- Department of Pharmacy, Xi'an Jiaotong University, Xi'an, 710003, China
| |
Collapse
|
5
|
Wolosowicz M, Prokopiuk S, Kaminski TW. The Complex Role of Matrix Metalloproteinase-2 (MMP-2) in Health and Disease. Int J Mol Sci 2024; 25:13691. [PMID: 39769454 PMCID: PMC11728377 DOI: 10.3390/ijms252413691] [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: 11/29/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Matrix metalloproteinase-2 (MMP-2), a zinc-dependent enzyme, plays a critical role in the degradation and remodeling of the extracellular matrix (ECM). As a member of the gelatinase subgroup of matrix metalloproteinases, MMP-2 is involved in a variety of physiological processes, including tissue repair, wound healing, angiogenesis, and embryogenesis. It is primarily responsible for the degradation of type IV and V collagen, fibronectin, laminin, and elastin, which are essential components of the ECM. MMP-2 is secreted as an inactive pro-enzyme (proMMP-2) and activated through proteolytic cleavage, with its activity being precisely regulated by tissue inhibitors of metalloproteinases (TIMPs). Dysregulation of MMP-2 has been linked to a variety of pathological conditions, including cardiovascular diseases, diabetic complications, kidney diseases, and cancer. In cardiovascular diseases, it contributes to vascular remodeling, atherosclerosis, and aneurysms, while in fibrotic diseases, it mediates excessive ECM degradation leading to tissue scarring. In diabetes, elevated MMP-2 activity exacerbates complications such as nephropathy, retinopathy, and cardiovascular disease. In cancer, MMP-2 facilitates tumor invasion and metastasis by degrading ECM components and promoting angiogenesis. Despite its essential roles in both physiological and pathological processes, targeting MMP-2 for therapeutic purposes presents challenges due to its dual functions in tissue remodeling and repair, raising concerns about unplanned consequences such as impaired tissue healing or excessive tissue damage. These challenges underscore the need for future research to focus on developing selective modulators that can precisely balance their activity under specific disease environments. Clinical trials targeting MMP-2 modulation highlight the potential of gelatinase inhibitors, including those targeting MMP-2, to reduce tumor progression in fibrosarcoma, breast, and lung cancers. This paper reviews the structure, function, and regulation of MMP-2, its involvement in disease pathogenesis, and the potential challenges in the therapeutic implications of modulating its activity.
Collapse
Affiliation(s)
- Marta Wolosowicz
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Slawomir Prokopiuk
- Faculty of Health Sciences, University of Lomza, 14 Akademicka St., 18-400 Łomża, Poland;
| | - Tomasz W. Kaminski
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute (VMI), University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
- Thrombosis and Hemostasis Program, VERSITI Blood Research Institute, Milwaukee, WI 53226, USA
| |
Collapse
|
6
|
Zahid R, Wang J, Cai Z, Ishtiaq A, Liu M, Ma D, Liang Y, Xu Y. Single chain fragment variable, a new theranostic approach for cardiovascular diseases. Front Immunol 2024; 15:1443290. [PMID: 39735545 PMCID: PMC11671482 DOI: 10.3389/fimmu.2024.1443290] [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: 06/03/2024] [Accepted: 11/26/2024] [Indexed: 12/31/2024] Open
Abstract
Cardiovascular diseases (CVDs) remain a significant global health challenge, leading to substantial morbidity and mortality. Despite recent advancements in CVD management, pharmaceutical treatments often suffer from poor pharmacokinetics and high toxicity. With the rapid progress of modern molecular biology and immunology, however, single-chain fragment variable (scFv) molecule engineering has emerged as a promising theranostic tool to offer specificity and versatility in targeting CVD-related antigens. To represent the latest development on the potential of scFv in the context of CVDs, this review summarized the new mechanism of action and applications as therapeutic, as well as diagnostic agents. Furthermore, the advantages of scFv, including its small size, ease of modification, and ability to be engineered for enhanced affinity and specificity, are also described. Finally, such challenges as immunogenicity, stability, and scalability, alongside strategies to overcome these hurdles, are deeply scrutinized to provide safer and more effective strategies for the diagnosis and treatment of the incurable CVDs.
Collapse
Affiliation(s)
- Rukhshan Zahid
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Juncheng Wang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Zecheng Cai
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Ayesha Ishtiaq
- College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Meng Liu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Dan Ma
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Yan Liang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| |
Collapse
|
7
|
Wang Y, Wang C, Li J. Neutrophil extracellular traps: a catalyst for atherosclerosis. Mol Cell Biochem 2024; 479:3213-3227. [PMID: 38401035 DOI: 10.1007/s11010-024-04931-3] [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: 10/23/2023] [Accepted: 01/05/2024] [Indexed: 02/26/2024]
Abstract
Neutrophil extracellular traps (NETs) are network-like structures released by activated neutrophils. They consist mainly of double-stranded DNA, histones, and neutrophil granule proteins. Continuous release of NETs in response to external stimuli leads to activation of surrounding platelets and monocytes/macrophages, resulting in damage to endothelial cells (EC) and vascular smooth muscle cells (VSMC). Some clinical trials have demonstrated the association between NETs and the severity and prognosis of atherosclerosis. Furthermore, experimental findings have shed light on the molecular mechanisms by which NETs contribute to atherogenesis. NETs play a significant role in the formation of atherosclerotic plaques. This review focuses on recent advancements in the understanding of the relationship between NETs and atherosclerosis. It explores various aspects, including the formation of NETs in atherosclerosis, clinical trials investigating NET-induced atherosclerosis, the mechanisms by which NETs promote atherogenesis, and the translational implications of NETs. Ultimately, we aim to propose new research directions for the diagnosis and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yinyu Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Cuiping Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Jiayan Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
8
|
Momi S, Gresele P. The Role of Platelets in Atherosclerosis: A Historical Review. Semin Thromb Hemost 2024. [PMID: 39561814 DOI: 10.1055/s-0044-1795097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Atherosclerosis is a chronic, multifactorial inflammatory disorder of large and medium-size arteries, which is the leading cause of cardiovascular mortality and morbidity worldwide. Although platelets in cardiovascular disease have mainly been studied for their crucial role in the thrombotic event triggered by atherosclerotic plaque rupture, over the last two decades it has become clear that platelets participate also in the development of atherosclerosis, owing to their ability to interact with the damaged arterial wall and with leukocytes. Platelets participate in all phases of atherogenesis, from the initial functional damage to endothelial cells to plaque unstabilization. Platelets deposit at atherosclerosis predilection sites before the appearance of manifest lesions to the endothelium and contribute to induce endothelial dysfunction, thus supporting leukocyte adhesion to the vessel wall. In particular, platelets release matrix metalloproteinases, which interact with protease-activated receptor 1 on endothelial cells triggering adhesion molecule expression. Moreover, P-selectin and glycoprotein Ibα expressed on the surface of vessel wall-adhering platelets bind PSGL-1 and β2 integrins on leukocytes, favoring their arrest and transendothelial migration. Platelet-leukocyte interactions promote the formation of radical oxygen species which are strongly involved in the lipid peroxidation associated with atherosclerosis. Platelets themselves actively migrate through the endothelium toward the plaque core where they release chemokines that modify the microenvironment by modulating the function of other inflammatory cells, such as macrophages. While current antiplatelet agents seem unable to prevent the contribution of platelets to atherogenesis, the inhibition of platelet secretion, of the release of MMPs, and of some specific pathways of platelet adhesion to the vessel wall may represent promising future strategies for the prevention of atheroprogression.
Collapse
Affiliation(s)
- Stefania Momi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paolo Gresele
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
9
|
Paciullo F, Falcinelli E, Fierro T, Gresele P, Del Pinto M. High - but not standard-dose atorvastatin prevents the increase of plasma matrix metalloproteinase-2 triggered by acute coronary syndromes. Coron Artery Dis 2024; 35:622-624. [PMID: 39087641 PMCID: PMC11426972 DOI: 10.1097/mca.0000000000001414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 07/13/2024] [Indexed: 08/02/2024]
Affiliation(s)
- Francesco Paciullo
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia
| | - Emanuela Falcinelli
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia
| | - Tiziana Fierro
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia
| | - Paolo Gresele
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia
| | | |
Collapse
|
10
|
Rodriguez L, Muñoz-Bernal ÓA, Fuentes E, Alvarez-Parrilla E, Palomo I, Wall-Medrano A. Phenolic profile, cheminformatics, and antiplatelet aggregation activity of orange and purple sweet potato (Ipomoea batatas L.) storage roots. Food Chem 2024; 454:139794. [PMID: 38797094 DOI: 10.1016/j.foodchem.2024.139794] [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: 02/16/2024] [Revised: 05/04/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
Sweet potatoes are rich in cardioprotective phytochemicals with potential anti-platelet aggregation activity, although this benefit may vary among cultivars/genotypes. The phenolic profile [HPLC-ESI(-)-qTOF-MS2], cheminformatics (ADMET properties, affinity toward platelet proteins) and anti-PA activity of phenolic-rich hydroalcoholic extracts obtained from orange (OSP) and purple (PSP) sweet potato storage roots, was evaluated. The phenolic richness [Hydroxycinnamic acids> flavonoids> benzoic acids] was PSP > OSP. Their main chlorogenic acids could interact with platelet proteins (integrins/adhesins, kinases/metalloenzymes) but their bioavailability could be poor. Just OSP exhibited a dose-dependent anti-platelet aggregation activity [inductor (IC50, mg.ml-1): thrombin receptor activator peptide-6 (0.55) > Adenosine-5'-diphosphate (1.02) > collagen (1.56)] and reduced P-selectin expression (0.75-1.0 mg.ml-1) but not glycoprotein IIb/IIIa secretion. The explored anti-PA activity of OSP/PSP seems to be inversely related to their phenolic richness. The poor first-pass bioavailability of its chlorogenic acids (documented in silico) may represent a further obstacle for their anti-PA in vivo.
Collapse
Affiliation(s)
- Lyanne Rodriguez
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Av. Lircay s/n, Talca 3460000, Chile.
| | - Óscar Adrian Muñoz-Bernal
- Departamento de Ciencias Químico-Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Anillo Envolvente del Pronaf y Estocolmo s/n, Fovisste Chamizal, Ciudad Juárez 32310, Chihuahua, Mexico.
| | - Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Av. Lircay s/n, Talca 3460000, Chile.
| | - Emilio Alvarez-Parrilla
- Departamento de Ciencias Químico-Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Anillo Envolvente del Pronaf y Estocolmo s/n, Fovisste Chamizal, Ciudad Juárez 32310, Chihuahua, Mexico.
| | - Iván Palomo
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Av. Lircay s/n, Talca 3460000, Chile.
| | - Abraham Wall-Medrano
- Departamento de Ciencias Químico-Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Anillo Envolvente del Pronaf y Estocolmo s/n, Fovisste Chamizal, Ciudad Juárez 32310, Chihuahua, Mexico.
| |
Collapse
|
11
|
Zhao K, Zeng Z, He Y, Zhao R, Niu J, Sun H, Li S, Dong J, Jing Z, Zhou J. Recent advances in targeted therapy for inflammatory vascular diseases. J Control Release 2024; 372:730-750. [PMID: 38945301 DOI: 10.1016/j.jconrel.2024.06.063] [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: 01/07/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
Vascular diseases constitute a significant contributor to worldwide mortality rates, placing a substantial strain on healthcare systems and socio-economic aspects. They are closely associated with inflammatory responses, as sustained inflammation could impact endothelial function, the release of inflammatory mediators, and platelet activation, thus accelerating the progression of vascular diseases. Consequently, directing therapeutic efforts towards mitigating inflammation represents a crucial approach in the management of vascular diseases. Traditional anti-inflammatory medications may have extensive effects on multiple tissues and organs when absorbed through the bloodstream. Conversely, treatments targeting inflammatory vascular diseases, such as monoclonal antibodies, drug-eluting stents, and nano-drugs, can achieve more precise effects, including precise intervention, minimal non-specific effects, and prolonged efficacy. In addition, personalized therapy is an important development trend in targeted therapy for inflammatory vascular diseases. Leveraging advanced simulation algorithms and clinical trial data, treatment strategies are gradually being personalized based on patients' genetic, biomarker, and clinical profiles. It is expected that the application of precision medicine in the field of vascular diseases will have a broader future. In conclusion, targeting therapies offer enhanced safety and efficacy compared to conventional medications; investigating novel targeting therapies and promoting clinical transformation may be a promising direction in improving the prognosis of patients with inflammatory vascular diseases. This article reviews the pathogenesis of inflammatory vascular diseases and presents a comprehensive overview of the potential for targeted therapies in managing this condition.
Collapse
Affiliation(s)
- Kaiwen Zhao
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Zan Zeng
- Department of Vascular Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Yuzhen He
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Rong Zhao
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Jinzhu Niu
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Huiying Sun
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Shuangshuang Li
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Jian Dong
- Department of Vascular Surgery, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zaiping Jing
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Jian Zhou
- Department of Vascular Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, China; Department of Vascular Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China; Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai, China.
| |
Collapse
|
12
|
Li S, He RC, Wu SG, Song Y, Zhang KL, Tang ML, Bei YR, Zhang T, Lu JB, Ma X, Jiang M, Qin LJ, Xu Y, Dong XH, Wu J, Dai X, Hu YW. LncRNA PSMB8-AS1 Instigates Vascular Inflammation to Aggravate Atherosclerosis. Circ Res 2024; 134:60-80. [PMID: 38084631 DOI: 10.1161/circresaha.122.322360] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 11/20/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Increasing evidence suggests that long noncoding RNAs play significant roles in vascular biology and disease development. One such long noncoding RNA, PSMB8-AS1, has been implicated in the development of tumors. Nevertheless, the precise role of PSMB8-AS1 in cardiovascular diseases, particularly atherosclerosis, has not been thoroughly elucidated. Thus, the primary aim of this investigation is to assess the influence of PSMB8-AS1 on vascular inflammation and the initiation of atherosclerosis. METHODS We generated PSMB8-AS1 knockin and Apoe (Apolipoprotein E) knockout mice (Apoe-/-PSMB8-AS1KI) and global Apoe and proteasome subunit-β type-9 (Psmb9) double knockout mice (Apoe-/-Psmb9-/-). To explore the roles of PSMB8-AS1 and Psmb9 in atherosclerosis, we fed the mice with a Western diet for 12 weeks. RESULTS Long noncoding RNA PSMB8-AS1 is significantly elevated in human atherosclerotic plaques. Strikingly, Apoe-/-PSMB8-AS1KI mice exhibited increased atherosclerosis development, plaque vulnerability, and vascular inflammation compared with Apoe-/- mice. Moreover, the levels of VCAM1 (vascular adhesion molecule 1) and ICAM1 (intracellular adhesion molecule 1) were significantly upregulated in atherosclerotic lesions and serum of Apoe-/-PSMB8-AS1KI mice. Consistently, in vitro gain- and loss-of-function studies demonstrated that PSMB8-AS1 induced monocyte/macrophage adhesion to endothelial cells and increased VCAM1 and ICAM1 levels in a PSMB9-dependent manner. Mechanistic studies revealed that PSMB8-AS1 induced PSMB9 transcription by recruiting the transcription factor NONO (non-POU domain-containing octamer-binding protein) and binding to the PSMB9 promoter. PSMB9 (proteasome subunit-β type-9) elevated VCAM1 and ICAM1 expression via the upregulation of ZEB1 (zinc finger E-box-binding homeobox 1). Psmb9 deficiency decreased atherosclerotic lesion size, plaque vulnerability, and vascular inflammation in Apoe-/- mice in vivo. Importantly, endothelial overexpression of PSMB8-AS1-increased atherosclerosis and vascular inflammation were attenuated by Psmb9 knockout. CONCLUSIONS PSMB8-AS1 promotes vascular inflammation and atherosclerosis via the NONO/PSMB9/ZEB1 axis. Our findings support the development of new long noncoding RNA-based strategies to counteract atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Shu Li
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Run-Chao He
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Shao-Guo Wu
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangdong, China (S.-G.W.)
| | - Yu Song
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Ke-Lan Zhang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Mao-Lin Tang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Yan-Rou Bei
- Laboratory Medicine Center (Y.-R.B.), Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ting Zhang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Jin-Bo Lu
- Department of Peripheral Vascular Surgery, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen (J.-B.L.)
| | - Xin Ma
- Department of Anesthesiology (X.M.), Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Min Jiang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Liang-Jun Qin
- Department of Pathology, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (L.J.Q.)
| | - Yudan Xu
- Laboratory Medicine Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China (Y.X.)
| | - Xian-Hui Dong
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Jia Wu
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Xiaoyan Dai
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangdong, China (X.D.)
- Clinical Research Institute, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, China (X.D.)
| | - Yan-Wei Hu
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
- Department of Laboratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (Y.-W.H.)
| |
Collapse
|
13
|
Crea F. The key role of thrombosis: focus on acute coronary syndrome, venous thrombo-embolism, and atrial fibrillation. Eur Heart J 2024; 45:1-4. [PMID: 38160711 DOI: 10.1093/eurheartj/ehad835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Affiliation(s)
- Filippo Crea
- Centre of Excellence of Cardiovascular Sciences, Gemelli Isola Hospital, Rome, Italy
| |
Collapse
|
14
|
Ye W, Wang J, Little PJ, Zou J, Zheng Z, Lu J, Yin Y, Liu H, Zhang D, Liu P, Xu S, Ye W, Liu Z. Anti-atherosclerotic effects and molecular targets of ginkgolide B from Ginkgo biloba. Acta Pharm Sin B 2024; 14:1-19. [PMID: 38239238 PMCID: PMC10792990 DOI: 10.1016/j.apsb.2023.09.014] [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: 06/08/2023] [Revised: 09/03/2023] [Accepted: 09/13/2023] [Indexed: 01/22/2024] Open
Abstract
Bioactive compounds derived from herbal medicinal plants modulate various therapeutic targets and signaling pathways associated with cardiovascular diseases (CVDs), the world's primary cause of death. Ginkgo biloba , a well-known traditional Chinese medicine with notable cardiovascular actions, has been used as a cardio- and cerebrovascular therapeutic drug and nutraceutical in Asian countries for centuries. Preclinical studies have shown that ginkgolide B, a bioactive component in Ginkgo biloba , can ameliorate atherosclerosis in cultured vascular cells and disease models. Of clinical relevance, several clinical trials are ongoing or being completed to examine the efficacy and safety of ginkgolide B-related drug preparations in the prevention of cerebrovascular diseases, such as ischemia stroke. Here, we present a comprehensive review of the pharmacological activities, pharmacokinetic characteristics, and mechanisms of action of ginkgolide B in atherosclerosis prevention and therapy. We highlight new molecular targets of ginkgolide B, including nicotinamide adenine dinucleotide phosphate oxidases (NADPH oxidase), lectin-like oxidized LDL receptor-1 (LOX-1), sirtuin 1 (SIRT1), platelet-activating factor (PAF), proprotein convertase subtilisin/kexin type 9 (PCSK9) and others. Finally, we provide an overview and discussion of the therapeutic potential of ginkgolide B and highlight the future perspective of developing ginkgolide B as an effective therapeutic agent for treating atherosclerosis.
Collapse
Affiliation(s)
- Weile Ye
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Jiaojiao Wang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Peter J. Little
- Pharmacy Australia Centre of Excellence, School of Pharmacy, University of Queensland, Woolloongabba QLD 4102, Australia
- Sunshine Coast Health Institute and School of Health and Behavioural Sciences, University of the Sunshine Coast, Birtinya QLD 4575, Australia
| | - Jiami Zou
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Zhihua Zheng
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Jing Lu
- National-Local Joint Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Yanjun Yin
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - Hao Liu
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - Dongmei Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Peiqing Liu
- National-Local Joint Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Suowen Xu
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
- Institute of Endocrine and Metabolic Diseases, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Wencai Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Zhiping Liu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| |
Collapse
|
15
|
Baaten CCFMJ, Nagy M, Bergmeier W, Spronk HMH, van der Meijden PEJ. Platelet biology and function: plaque erosion vs. rupture. Eur Heart J 2024; 45:18-31. [PMID: 37940193 PMCID: PMC10757869 DOI: 10.1093/eurheartj/ehad720] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/20/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
The leading cause of heart disease in developed countries is coronary atherosclerosis, which is not simply a result of ageing but a chronic inflammatory process that can lead to acute clinical events upon atherosclerotic plaque rupture or erosion and arterial thrombus formation. The composition and location of atherosclerotic plaques determine the phenotype of the lesion and whether it is more likely to rupture or to erode. Although plaque rupture and erosion both initiate platelet activation on the exposed vascular surface, the contribution of platelets to thrombus formation differs between the two phenotypes. In this review, plaque phenotype is discussed in relation to thrombus composition, and an overview of important mediators (haemodynamics, matrix components, and soluble factors) in plaque-induced platelet activation is given. As thrombus formation on disrupted plaques does not necessarily result in complete vessel occlusion, plaque healing can occur. Therefore, the latest findings on plaque healing and the potential role of platelets in this process are summarized. Finally, the clinical need for more effective antithrombotic agents is highlighted.
Collapse
Affiliation(s)
- Constance C F M J Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Magdolna Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, School of Medicine, University of North Caroline at Chapel Hill, Chapel Hill, NC, USA
- Blood Research Center, School of Medicine, University of North Caroline at Chapel Hill, Chapel Hill, NC, USA
| | - Henri M H Spronk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
- Thrombosis Expertise Center, Heart+ Vascular Center, Maastricht University Medical Center+, P. Debeyelaan 25, Maastricht, the Netherlands
| | - Paola E J van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- Thrombosis Expertise Center, Heart+ Vascular Center, Maastricht University Medical Center+, P. Debeyelaan 25, Maastricht, the Netherlands
| |
Collapse
|
16
|
Gao Y, Li Y, Chen X, Wu C, Guo Z, Bai G, Liu T, Li G. The Systemic Inflammation Index Predicts Poor Clinical Prognosis in Patients with Initially Diagnosed Acute Coronary Syndrome Undergoing Primary Coronary Angiography. J Inflamm Res 2023; 16:5205-5219. [PMID: 38026253 PMCID: PMC10655605 DOI: 10.2147/jir.s435398] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Systemic inflammation index (SII: neutrophil count * platelet count/lymphocyte count) is a new inflammatory marker that can reflect the degree of systemic inflammatory response after coronary artery disease (CAD). However, the predictive value of the SII for clinical prognosis in patients with initially diagnosed acute coronary syndrome (ACS) has yet to be thoroughly studied. PATIENTS AND METHODS Patients with initially diagnosed ACS who underwent primary coronary angiography in our hospital from January 2019 to April 2021 were included in this study. 757 patients with ACS who underwent primary coronary angiography were enrolled. According to the baseline SII level, the patients were divided into a high SII group and a low SII group. The primary endpoint was major cardiovascular events (MACEs), defined as cardiac death, non-fatal myocardial infarction (MI), and non-fatal stroke. RESULTS At a median follow-up of 33.9 months, 140 (18.5%) MACEs were recorded. Receiver operating characteristic (ROC) curve analysis showed that SII's best cut-off value for predicting MACEs was 713.9*109/L. Kaplan-Meier survival curve analysis showed that the survival rate of the low SII group was higher than the high SII group (P<0.001). Compared with the low SII group, the risk of MACEs was significantly increased in the high SII group (89 cases (33.3%) vs.51 patients (10.4%), P<0.001). Univariate and multivariate Cox regression analysis manifested that high SII level was independently associated with the occurrence of MACEs in patients with ACS undergoing primary coronary angiography (adjusted hazard ratio [HR]: 2.915, 95% confidence interval (CI%): 1.830-4.641, P<0.001). Adding SII to the conventional risk factor model improved the predictive value of MACEs. CONCLUSION This study showed that elevated SII was associated with adverse cardiovascular prognosis in patients with ACS undergoing primary coronary angiography, making SII a valuable predictor of poor prognosis in patients with ACS undergoing primary coronary angiography.
Collapse
Affiliation(s)
- Yi Gao
- Tianjin Key Laboratory of Logic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Yuqing Li
- Tianjin Key Laboratory of Logic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Xiaolin Chen
- Tianjin Key Laboratory of Logic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Chen Wu
- Department of Emergency Medicine, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Ziqiang Guo
- Tianjin Key Laboratory of Logic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Geng Bai
- Tianjin Key Laboratory of Logic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Tong Liu
- Tianjin Key Laboratory of Logic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Guangping Li
- Tianjin Key Laboratory of Logic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| |
Collapse
|
17
|
Buckley LF, Agha AM, Dorbala P, Claggett BL, Yu B, Hussain A, Nambi V, Chen LY, Matsushita K, Hoogeveen RC, Ballantyne CM, Shah AM. MMP-2 Associates With Incident Heart Failure and Atrial Fibrillation: The ARIC Study. Circ Heart Fail 2023; 16:e010849. [PMID: 37753653 PMCID: PMC10842537 DOI: 10.1161/circheartfailure.123.010849] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/16/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND MMP (matrix metalloproteinase)-2 participates in extracellular matrix regulation and may be involved in heart failure (HF), atrial fibrillation (AF), and coronary heart disease. METHODS Among the 4693 ARIC study (Atherosclerosis Risk in Communities) participants (mean age, 75±5 years; 42% women) without prevalent HF, multivariable Cox proportional hazard models were used to estimate associations of plasma MMP-2 levels with incident HF, HF with preserved ejection fraction (≥50%), HF with reduced ejection fraction (<50%), AF, and coronary heart disease. Mediation of the association between MMP-2 and HF was assessed by censoring participants who developed AF or coronary heart disease before HF. Multivariable linear regression models were used to assess associations of MMP-2 with measures of left ventricular and left atrial structure and function. RESULTS Compared with the 3 lower quartiles, the highest MMP-2 quartile associated with greater risk of incident HF overall (adjusted hazard ratio, 1.48 [95% CI, 1.21-1.81]), incident HF with preserved ejection fraction (1.44 [95% CI, 1.07-1.94]), incident heart failure with reduced ejection fraction (1.48 [95% CI, 1.08-2.02]), and incident AF (1.44 [95% CI, 1.18-1.77]) but not incident coronary heart disease (0.97 [95% CI, 0.71-1.34]). Censoring AF attenuated the MMP-2 association with HF with preserved ejection fraction. Higher plasma MMP-2 levels were associated with larger left ventricular end-diastolic volume index, greater left ventricular mass index, higher E/e' ratio, larger left atrial volume index, and worse left atrial reservoir and contractile strains (all P<0.001). CONCLUSIONS Higher plasma MMP-2 levels associate with diastolic dysfunction, left atrial dysfunction, and a higher risk of incident HF and AF. AF is a mediator of MMP-2-associated HF with preserved ejection fraction risk.
Collapse
Affiliation(s)
- Leo F Buckley
- Department of Pharmacy Services (L.F.B.), Brigham and Women's Hospital, Boston, MA
| | - Ali M Agha
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
| | - Pranav Dorbala
- Division of Cardiovascular Medicine (P.D., B.L.C.), Brigham and Women's Hospital, Boston, MA
| | - Brian L Claggett
- Division of Cardiovascular Medicine (P.D., B.L.C.), Brigham and Women's Hospital, Boston, MA
| | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas Health Science Center at Houston (B.Y.)
| | - Aliza Hussain
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
| | - Vijay Nambi
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
- Michael E. DeBakey Veterans Affairs Hospital, Houston, TX (V.N.)
| | - Lin Yee Chen
- Division of Cardiovascular Medicine, University of Minnesota, Minneapolis (L.Y.C.)
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.)
| | - Ron C Hoogeveen
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
| | - Christie M Ballantyne
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
| | - Amil M Shah
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas (A.M.S.)
| |
Collapse
|
18
|
Rao S, Grover M. Intestinal proteases. Curr Opin Gastroenterol 2023; 39:472-478. [PMID: 37678185 PMCID: PMC10592107 DOI: 10.1097/mog.0000000000000972] [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] [Indexed: 09/09/2023]
Abstract
PURPOSE OF REVIEW Proteases constitute a group of enzymes that hydrolyze peptide bonds. Intestinal proteases are an integral part of gut homeostasis and digestion. This review discusses the broader classification of proteases, regulation of proteolytic activity (PA) in the intestinal tract, and how dysregulation of intestinal proteases contributes to the pathophysiology of conditions such as irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and celiac disease. We also discuss recent advancements in therapeutic modulation that directly or indirectly target intestinal proteases and can be utilized to treat these illnesses. RECENT FINDINGS Host and microbiota derived proteases have been associated with symptoms in subsets of patients with IBS, IBD and celiac disease. Elevated PA mediates barrier dysfunction, visceral hypersensitivity as well as immune activation and inflammation. Recent mechanistic studies have revealed the nature of disease-associated proteases and mechanisms regulating their activity, particularly those driven by the microbiota. Advancements in activity-based probes have allowed novel ways of in vivo imaging of PA. Newer strategies targeting proteases include monoclonal antibodies, engineered microbiota as well as specific protease inhibitors. SUMMARY Significant progresses made in the detection as well as regulation of PA is likely to provide therapeutic advancements for gastrointestinal diseases.
Collapse
Affiliation(s)
- Sameer Rao
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
- Sawai Man Singh Medical College, Jaipur, India
| | - Madhusudan Grover
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
19
|
Samah N, Ugusman A, Hamid AA, Sulaiman N, Aminuddin A. Role of Matrix Metalloproteinase-2 in the Development of Atherosclerosis among Patients with Coronary Artery Disease. Mediators Inflamm 2023; 2023:9715114. [PMID: 37457745 PMCID: PMC10348858 DOI: 10.1155/2023/9715114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
Coronary artery disease (CAD) is a caused by atherosclerotic plaque buildup in the coronary arteries that supply blood and oxygen to the heart. Matrix metalloproteinase (MMP) is a family of zinc-dependent endopeptidase that is involved in various stages of atherosclerosis as demonstrated in in vitro and in vivo studies. MMP-2 is associated with both stable and unstable atherosclerotic plaque formation. The current review aimed to identify the role of MMP-2 in atherosclerosis development among CAD patients. Literature search was conducted through four online databases and only studies that were published from 2018 until February 2023 were included. The risk of bias was assessed by using the Newcastle-Ottawa Scale. A total of 10,622 articles were initially identified, and only eight studies that fulfilled the selection criteria were included in this review. The results showed that MMP-2 levels and activity were higher in patients with unstable CAD than those with stable CAD and healthy subjects. There was a significant association between MMP-2 levels and cardiovascular disease with MMP-14 levels, which is a pro-MMP-2 activator. In addition, two single nucleotide polymorphisms of the MMP-2 gene (rs243865 and rs243866) were significantly associated with the development of atherosclerosis. In conclusion, MMP-2 plays a crucial role in the development of atherosclerosis among patients with CAD and could be a potential target for CAD therapy.
Collapse
Affiliation(s)
- Nazirah Samah
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Adila A. Hamid
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Nadiah Sulaiman
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Amilia Aminuddin
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
20
|
Ju Y, Choi GE, Lee MW, Jeong M, Kwon H, Kim DH, Kim J, Jin H, Lee KE, Hyun KY, Jang A. Identification of miR-143-3p as a diagnostic biomarker in gastric cancer. BMC Med Genomics 2023; 16:135. [PMID: 37328880 PMCID: PMC10273760 DOI: 10.1186/s12920-023-01554-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/19/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is among the most common types of gastrointestinal cancers and has a high incidence and mortality around the world. To suppress the progression of GC, it is essential to develop diagnostic markers. MicroRNAs regulate GC development, but a clearer insight into their role is needed before they can be applied as a molecular markers and targets. METHODS In this study, we assessed the diagnostic value of differentially expressed microRNAs as potential diagnostic biomarkers for GC using data for 389 tissue samples from the Cancer Genome Atlas (TCGA) and 21 plasma samples from GC patients. RESULTS The expression of hsa-miR-143-3p (also known as hsa-miR-143) was significantly downregulated in GC according to the TCGA data and plasma samples. The 228 potential target genes of hsa-miR-143-3p were analyzed using a bioinformatics tool for miRNA target prediction. The target genes correlated with extracellular matrix organization, the cytoplasm, and identical protein binding. Furthermore, the pathway enrichment analysis of target genes showed that they were involved in pathways in cancer, the phosphoinositide 3-kinase (PI3K)-protein kinase B (Akt) signaling pathway, and proteoglycans in cancer. The hub genes in the protein-protein interaction (PPI) network, were matrix metallopeptidase 2 (MMP2), CD44 molecule (CD44), and SMAD family member 3 (SMAD3). CONCLUSIONS This study suggests that hsa-miR-143-3p may be used as a diagnostic marker for GC, contributing via the pathways involved in the development of GC.
Collapse
Affiliation(s)
- Yeongdon Ju
- Medical Science Research Center, Pusan National University, Yangsan, 50612, Republic of Korea
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
| | - Go-Eun Choi
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
| | - Moon Won Lee
- Division of Gastroenterology, Pusan National University Hospital, Busan, 49241, Republic of Korea
- Department of Internal Medicine, Pusan National University College of Medicine, Busan, 49241, Republic of Korea
| | - Myeongguk Jeong
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
| | - Hyeokjin Kwon
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
| | - Dong Hyeok Kim
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
| | - Jungho Kim
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
| | - Hyunwoo Jin
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
| | - Kyung Eun Lee
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
| | - Kyung-Yae Hyun
- Department of Clinical Laboratory Science, Dong-Eui University, Busan, 47340, Republic of Korea.
| | - Aelee Jang
- Department of Nursing, University of Ulsan, Ulsan, 44610, Republic of Korea.
| |
Collapse
|
21
|
Crea F. Thrombotic and bleeding complications during antithrombotic treatment: the need for new therapeutic targets. Eur Heart J 2023; 44:1767-1770. [PMID: 37210634 DOI: 10.1093/eurheartj/ehad285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/22/2023] Open
Affiliation(s)
- Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
22
|
Rolling CC, Barrett TJ, Berger JS. Platelet-monocyte aggregates: molecular mediators of thromboinflammation. Front Cardiovasc Med 2023; 10:960398. [PMID: 37255704 PMCID: PMC10225702 DOI: 10.3389/fcvm.2023.960398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 04/24/2023] [Indexed: 06/01/2023] Open
Abstract
Platelets, key facilitators of primary hemostasis and thrombosis, have emerged as crucial cellular mediators of innate immunity and inflammation. Exemplified by their ability to alter the phenotype and function of monocytes, activated platelets bind to circulating monocytes to form monocyte-platelet aggregates (MPA). The platelet-monocyte axis has emerged as a key mechanism connecting thrombosis and inflammation. MPA are elevated across the spectrum of inflammatory and autoimmune disorders, including cardiovascular disease, systemic lupus erythematosus (SLE), and COVID-19, and are positively associated with disease severity. These clinical disorders are all characterized by an increased risk of thromboembolic complications. Intriguingly, monocytes in contact with platelets become proinflammatory and procoagulant, highlighting that this interaction is a central element of thromboinflammation.
Collapse
Affiliation(s)
- Christina C. Rolling
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tessa J. Barrett
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Jeffrey S. Berger
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
23
|
Crea F. Ischaemic heart disease: antithrombotic treatment and new therapeutic targets. Eur Heart J 2023; 44:239-242. [PMID: 36680421 DOI: 10.1093/eurheartj/ehac816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
24
|
Bordeianu G, Mitu I, Stanescu RS, Ciobanu CP, Petrescu-Danila E, Marculescu AD, Dimitriu DC. Circulating Biomarkers for Laboratory Diagnostics of Atherosclerosis-Literature Review. Diagnostics (Basel) 2022; 12:diagnostics12123141. [PMID: 36553147 PMCID: PMC9777004 DOI: 10.3390/diagnostics12123141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is still considered a disease burden with long-term damaging processes towards the cardiovascular system. Evaluation of atherosclerotic stages requires the use of independent markers such as those already considered traditional, that remain the main therapeutic target for patients with atherosclerosis, together with emerging biomarkers. The challenge is finding models of predictive markers that are particularly tailored to detect and evaluate the evolution of incipient vascular lesions. Important advances have been made in this field, resulting in a more comprehensible and stronger linkage between the lipidic profile and the continuous inflammatory process. In this paper, we analysed the most recent data from the literature studying the molecular mechanisms of biomarkers and their involvement in the cascade of events that occur in the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
| | - Ivona Mitu
- Correspondence: (I.M.); (R.S.S.); Tel.: +40-75206-1747 (I.M.)
| | | | | | | | | | | |
Collapse
|
25
|
Novel approaches to antiplatelet therapy. Biochem Pharmacol 2022; 206:115297. [DOI: 10.1016/j.bcp.2022.115297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/05/2022] [Accepted: 10/05/2022] [Indexed: 11/20/2022]
|
26
|
Nagy M, van der Meijden PEJ, Glunz J, Schurgers L, Lutgens E, ten Cate H, Heitmeier S, Spronk HMH. Integrating Mechanisms in Thrombotic Peripheral Arterial Disease. Pharmaceuticals (Basel) 2022; 15:1428. [PMID: 36422558 PMCID: PMC9695058 DOI: 10.3390/ph15111428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/10/2023] Open
Abstract
Peripheral arterial disease (PAD), a manifestation of systemic atherosclerosis, is underdiagnosed in the general population. Despite the extensive research performed to unravel its pathophysiology, inadequate knowledge exists, thus preventing the development of new treatments. This review aims to highlight the essential elements of atherosclerosis contributing to the pathophysiology of PAD. Furthermore, emphasis will be placed on the role of thrombo-inflammation, with particular focus on platelet and coagulation activation as well as cell-cell interactions. Additional insight will be then discussed to reveal the contribution of hypercoagulability to the development of vascular diseases such as PAD. Lastly, the current antithrombotic treatments will be discussed, and light will be shed on promising new targets aiming to aid the development of new treatments.
Collapse
Affiliation(s)
- Magdolna Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Paola E. J. van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
| | - Julia Glunz
- Cardiovascular Research, Bayer AG, 42117 Wuppertal, Germany
| | - Leon Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Esther Lutgens
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 10785 Munich, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian’s University, 80539 Munich, Germany
- Experimental Cardiovascular Immunology Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Hugo ten Cate
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
- Department of Internal Medicine, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
- Center for Thrombosis and Hemostasis, Gutenberg University Mainz, 55122 Mainz, Germany
| | | | - Henri M. H. Spronk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
- Department of Internal Medicine, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
| |
Collapse
|
27
|
Degjoni A, Campolo F, Stefanini L, Venneri MA. The NO/cGMP/PKG pathway in platelets: The therapeutic potential of PDE5 inhibitors in platelet disorders. J Thromb Haemost 2022; 20:2465-2474. [PMID: 35950928 PMCID: PMC9805178 DOI: 10.1111/jth.15844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 01/09/2023]
Abstract
Platelets are the "guardians" of the blood circulatory system. At sites of vessel injury, they ensure hemostasis and promote immunity and vessel repair. However, their uncontrolled activation is one of the main drivers of thrombosis. To keep circulating platelets in a quiescent state, the endothelium releases platelet antagonists including nitric oxide (NO) that acts by stimulating the intracellular receptor guanylyl cyclase (GC). The latter produces the second messenger cyclic guanosine-3',5'-monophosphate (cGMP) that inhibits platelet activation by stimulating protein kinase G, which phosphorylates hundreds of intracellular targets. Intracellular cGMP pools are tightly regulated by a fine balance between GC and phosphodiesterases (PDEs) that are responsible for the hydrolysis of cyclic nucleotides. Phosphodiesterase type 5 (PDE5) is a cGMP-specific PDE, broadly expressed in most tissues in humans and rodents. In clinical practice, PDE5 inhibitors (PDE5i) are used as first-line therapy for erectile dysfunction, pulmonary artery hypertension, and lower urinary tract symptoms. However, several studies have shown that PDE5i may ameliorate the outcome of various other conditions, like heart failure and stroke. Interestingly, NO donors and cGMP analogs increase the capacity of anti-platelet drugs targeting the purinergic receptor type Y, subtype 12 (P2Y12) receptor to block platelet aggregation, and preclinical studies have shown that PDE5i inhibits platelet functions. This review summarizes the molecular mechanisms underlying the effect of PDE5i on platelet activation and aggregation focusing on the therapeutic potential of PDE5i in platelet disorders, and the outcomes of a combined therapy with PDE5i and NO donors to inhibit platelet activation.
Collapse
Affiliation(s)
- Anisa Degjoni
- Department of Experimental MedicineSapienza University of RomeRomeItaly
| | - Federica Campolo
- Department of Experimental MedicineSapienza University of RomeRomeItaly
| | - Lucia Stefanini
- Department of Translational and Precision MedicineSapienza University of RomeRomeItaly
| | - Mary Anna Venneri
- Department of Experimental MedicineSapienza University of RomeRomeItaly
| |
Collapse
|
28
|
Sun Y, Xu M, Wang C, Guan S, Wang L, Cong B, Zhu W, Xu Y. Low-molecular-weight fucoidan bidirectionally regulates lipid uptake and cholesterol efflux through the p38 MAPK phosphorylation. Int J Biol Macromol 2022; 220:371-384. [PMID: 35970372 DOI: 10.1016/j.ijbiomac.2022.08.059] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/26/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022]
Abstract
Atherosclerosis (AS) is the pathological basis of many cardiovascular and cerebrovascular diseases, in which macrophage-derived foam cells are the critical step and a typical pathological feature of early atherosclerosis. We previously confirmed that low-molecular-weight fucoidan (LMWF) had a good anti-AS effect, but the mechanism is still unclear. Here with aim to investigate the inhibitory effect of LMWF on foam cells and its molecular mechanism. Oil red O staining showed that LMWF effectively alleviated lipid accumulation and the formation of foam cells. Flow cytometry detection showed that LMWF promoted foam cells apoptosis. In addition, immunofluorescence showed that LMWF inhibited macrophage scavenger receptor A1 (SR-A1)-mediated lipid uptake and promoted ATP-binding cassette transporter A1 (ABCA1)-mediated cholesterol outflow. Western blot showed that LMWF downregulated SR-A1 protein expression and upregulated ABCA1 protein expression by inhibiting p38 mitogen activated protein kinase (p38MAPK) phosphorylation. Moreover, the mRNA transcriptions of Stat1, Elk-1, and Myc were downregulated when treated with LMWF. It concluded that, LMWF achieved bidirectional regulation of SR-A1 and ABCA1, then prevented the formation of foam cells, finally ameliorated the development of AS.
Collapse
Affiliation(s)
- Yu Sun
- Medical College, Qingdao University, Qingdao 266071, China
| | - Ming Xu
- Medical College, Qingdao University, Qingdao 266071, China
| | - Changxin Wang
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Shulong Guan
- Department of Surgery, Qingdao Shinan District People's Hospital, Qingdao 266520, China
| | - Lina Wang
- Department of Blood Transfusion, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shangdong University, Qingdao 266035, China
| | - Beibei Cong
- Central Laboratory, Qingdao Stomatological Hospital, Qingdao 266001, China.
| | - Wenlong Zhu
- Business School, Qingdao University of Technology, Qingdao 266520, China.
| | - Yingjie Xu
- Central Laboratory, Qingdao Stomatological Hospital, Qingdao 266001, China.
| |
Collapse
|
29
|
Falcinelli E, Petito E, Gresele P. The role of platelets, neutrophils and endothelium in COVID-19 infection. Expert Rev Hematol 2022; 15:727-745. [PMID: 35930267 DOI: 10.1080/17474086.2022.2110061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION COVID-19 is associated to an increased risk of thrombosis, as a result of a complex process that involves the activation of vascular and circulating cells, the release of soluble inflammatory and thrombotic mediators and blood clotting activation. AREAS COVERED This article reviews the pathophysiological role of platelets, neutrophils and the endothelium, and of their interactions, in the thrombotic complications of COVID-19 patients, and the current and future therapeutic approaches targeting these cell types. EXPERT OPINION Virus-induced platelet, neutrophil and endothelial cell changes are crucial triggers of the thrombotic complications and of the adverse evolution of COVID-19. Both the direct interaction with the virus and the associated cytokine storm concur to trigger cell activation in a classical thromboinflammatory vicious circle. Although heparin has proven to be an effective prophylactic and therapeutic weapon for the prevention and treatment of COVID-19-associated thrombosis, it acts downstream of the cascade of events triggered by SARS-CoV-2. The identification of specific molecular targets interrupting the thromboinflammatory cascade upstream, and more specifically acting either on the interaction of SARS-CoV-2 with blood and vascular cells or on the specific signalling mechanisms associated with their COVID-19-associated activation, might theoretically offer greater protection with potentially lesser side effects.
Collapse
Affiliation(s)
- E Falcinelli
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - E Petito
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - P Gresele
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
30
|
Sabetta A, Lombardi L, Stefanini L. Sex differences at the platelet-vascular interface. Intern Emerg Med 2022; 17:1267-1276. [PMID: 35576047 PMCID: PMC9352612 DOI: 10.1007/s11739-022-02994-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/16/2022] [Indexed: 12/24/2022]
Abstract
Platelets are multifunctional cells that ensure the integrity of the vascular wall and modulate the immune response at the blood/vascular interface. Their pathological activation results in both thrombosis and inflammation and implicates them in the pathogenesis of vascular disease. Vascular diseases are sexually dimorphic in terms of incidence, clinical presentation, outcome, and efficacy of anti-platelet therapy. We here provide an overview of what is known about the role of platelets in the initiation and progression of vascular diseases and summarize what is known about the sex differences in platelet reactivity and in the thromboinflammatory mechanisms that drive these diseases, with a particular focus on atherosclerosis, obstructive and non-obstructive coronary artery disease, and ischemic stroke. Understanding the sex differences at the platelet-vascular interface is clinically relevant as it will enable: (1) to design new therapeutic strategies that prevent the detrimental effects of the immune-modulatory function of platelets taking sex into account, and (2) to evaluate if sex-specific anti-platelet drug regimens should be used to reduce the risk not only of thrombosis but also of vascular disease progression.
Collapse
Affiliation(s)
- Annamaria Sabetta
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell'Università, 37, 00185, Rome, Italy
| | - Ludovica Lombardi
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell'Università, 37, 00185, Rome, Italy
| | - Lucia Stefanini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell'Università, 37, 00185, Rome, Italy.
| |
Collapse
|
31
|
Falcinelli E, De Paolis M, Boschetti E, Gresele P. Release of MMP-2 in the circulation of patients with acute coronary syndromes undergoing percutaneous coronary intervention: Role of platelets. Thromb Res 2022; 216:84-89. [PMID: 35759818 DOI: 10.1016/j.thromres.2022.06.006] [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: 04/12/2022] [Revised: 06/08/2022] [Accepted: 06/17/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Matrix metalloproteinases (MMPs) of atherosclerotic tissue contribute to plaque rupture triggering acute coronary syndromes (ACS). Several MMPs, including MMP-2, are also contained in platelets and released upon activation. An increase in circulating levels of MMP-2 has been reported in patients undergoing percutaneous coronary interventions (PCI), but its time-course and origin remain unclear. Aims of our study were to assess the time-course of MMP-2 release in blood of stable and unstable coronary artery disease patients undergoing PCI and to unravel the possible contribution of platelets to its release. METHODS Peripheral blood samples were drawn immediately before, 4 and 24 h after PCI from patients with ACS (NSTEMI or STEMI, n = 21) or with stable angina (SA, n = 21). Platelet-poor plasma and washed platelet lysates were prepared and stored for subsequent assay of MMP-2 and β-thromboglobulin (β-TG), a platelet-specific protein released upon activation. RESULTS Plasma MMP-2 and β-TG increased significantly 4 h after PCI and returned to baseline at 24 h in ACS patients, while they did not change in SA patients. Platelet content of MMP-2 and β-TG decreased significantly 4 h after PCI in patients with ACS, compatible with intravascular platelet activation and release, while they did not change in patients with SA. CONCLUSIONS PCI triggers the release of MMP-2 in the circulation of ACS patients but not in that of patients with SA. Platelets activated by PCI contribute to the increase of plasma MMP-2 releasing their MMP-2 content. Given that previous mechanicistic studies have shown that MMP-2 may sustain platelet activation and unstabilize downstream-located plaques and in the long term favour restenosis and atherosclerosis progression, these data may encourage the search for therapeutic agents blocking MMP-2 release or activity in ACS.
Collapse
Affiliation(s)
- Emanuela Falcinelli
- Department of Medicine and Surgery, Division of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Marcella De Paolis
- Department of Interventional Cardiology, Division of Cardiology, Santa Maria University Hospital, Terni, Italy
| | - Enrico Boschetti
- Department of Interventional Cardiology, Division of Cardiology, Santa Maria University Hospital, Terni, Italy
| | - Paolo Gresele
- Department of Medicine and Surgery, Division of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy.
| |
Collapse
|
32
|
Crea F. New therapeutic targets to reduce inflammation-associated cardiovascular risk: the CCL2-CCR2 axis, LOX-1, and IRF5. Eur Heart J 2022; 43:1777-1781. [PMID: 35567553 DOI: 10.1093/eurheartj/ehac233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
33
|
Crea F. New therapeutic targets in the prevention of atherosclerotic cardiovascular disease. Eur Heart J 2022; 43:435-439. [PMID: 35143652 DOI: 10.1093/eurheartj/ehac014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
34
|
Crea F. New targets in vascular biology and new challenges in vascular medicine. Eur Heart J 2021; 42:4285-4289. [PMID: 34743214 DOI: 10.1093/eurheartj/ehab747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
35
|
Affiliation(s)
- Judith M E M Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|