1
|
Kaur M, Aran KR, Paswan R. A potential role of gut microbiota in stroke: mechanisms, therapeutic strategies and future prospective. Psychopharmacology (Berl) 2024; 241:2409-2430. [PMID: 39463207 DOI: 10.1007/s00213-024-06708-4] [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: 03/19/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
RATIONALE Neurological conditions like Stroke and Alzheimer's disease (AD) often include inflammatory responses in the nervous system. Stroke, linked to high disability and mortality rates, poses challenges related to organ-related complications. Recent focus on understanding the pathophysiology of ischemic stroke includes aspects like cellular excitotoxicity, oxidative stress, cell death mechanisms, and neuroinflammation. OBJECTIVE The objective of this paper is to summarize and explore the pathophysiology of ischemic stroke, elucidates the gut-brain axis mechanism, and discusses recent clinical trials, shedding light on novel treatments and future possibilities. RESULTS Changes in gut architecture and microbiota contribute to dementia by enhancing intestinal permeability, activating the immune system, elevating proinflammatory mediators, altering blood-brain barrier (BBB) permeability, and ultimately leading to neurodegenerative diseases (NDDs). The gut-brain axis's potential role in disease pathophysiology offers new avenues for cell-based regenerative medicine in treating neurological conditions. CONCLUSION In conclusion, the gut microbiome significantly impacts stroke prognosis by highlighting the role of the gut-brain axis in ischemic stroke mechanisms. This insight suggests potential therapeutic strategies for improving outcomes.
Collapse
Affiliation(s)
- Manpreet Kaur
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| | - Raju Paswan
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
2
|
Liu CH, Rethi L, Weng PW, Trung Nguyen H, Chuang AEY. Cutting-edge advances in nano/biomedicine: A review on transforming thrombolytic therapy. Biochem Pharmacol 2024; 229:116523. [PMID: 39251141 DOI: 10.1016/j.bcp.2024.116523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
Thrombotic blockages within blood vessels give rise to critical cardiovascular disorders, including ischemic stroke, venous thromboembolism, and myocardial infarction. The current approach to the therapy of thrombolysis involves administering Plasminogen Activators (PA), but it is hindered by fast drug elimination, narrow treatment window, and the potential for bleeding complications. Leveraging nanomedicine to encapsulate and deliver PA offers a solution by improving the efficacy of therapy, safeguarding the medicine from proteinase biodegradation, and reducing unwanted effects in in vivo trials. In this review, we delve into the underlying venous as well as arterial thrombus pathophysiology and provide an overview of clinically approved PA used to address acute thrombotic conditions. We explore the existing challenges and potential directions within recent pivotal research on a variety of targeted nanocarriers, such as lipid, polymeric, inorganic, and biological carriers, designed for precise delivery of PA to specific sites. We also discuss the promising role of microbubbles and ultrasound-assisted Sono thrombolysis, which have exhibited enhanced thrombolysis in clinical studies. Furthermore, our review delves into approaches for the strategic development of nano-based carriers tailored for targeting thrombolytic action and efficient encapsulation of PA, considering the intricate interaction in biology systems as well as nanomaterials. In conclusion, the field of nanomedicine offers a valuable method for the exact and effective therapy of severe thrombus conditions, presenting a pathway toward improved patient outcomes and reduced complications.
Collapse
Affiliation(s)
- Chia-Hung Liu
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei 11031, Taiwan; TMU Research Center of Urology and Kidney, Taipei Medical University, 250 Wu-Hsing Street, Taipei 11031, Taiwan; Department of Urology, Shuang Ho Hospital, Taipei Medical University, 291 Zhongzheng Road, Zhonghe District, New Taipei City 23561, Taiwan
| | - Lekshmi Rethi
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Pei-Wei Weng
- Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Hieu Trung Nguyen
- Department of Orthopedics and Trauma, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Viet Nam
| | - Andrew E-Y Chuang
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei 11696, Taiwan.
| |
Collapse
|
3
|
Qureshi Z, Altaf F, Jamil A, Siddique R, Shah S. Breaking boundaries: exploring recent advances in anticoagulation and thrombosis management: a comprehensive review. Ann Med Surg (Lond) 2024; 86:6585-6597. [PMID: 39525737 PMCID: PMC11543160 DOI: 10.1097/ms9.0000000000002589] [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: 05/21/2024] [Accepted: 09/12/2024] [Indexed: 11/16/2024] Open
Abstract
Background Thromboembolic disorders globally contribute to morbidity and mortality, emphasizing adequate anticoagulation and thrombosis management. Therapeutic advances are essential in preventing complications like pulmonary embolism, stroke, and myocardial infarction. This review summarizes recent anticoagulation advances, current challenges, future directions, and novel anticoagulants and drug delivery systems on clinical outcomes. Methods This paper assesses the effectiveness and safety of new anticoagulants through a systematic review of recent clinical trials, meta-analyses, and guideline publications. Key studies, including PACIFIC-AF, RIVER, ENAVLE, ENVISAGE-TAVI AF, and ARCADIA, were analyzed to provide a perspective on therapeutic advancements. Results The review highlights key findings from vital clinical trials. Asundexian, in the PACIFIC-AF trial, demonstrated a 34% reduction in bleeding events compared to Apixaban. In the RIVER trial, Rivaroxaban reduced significant bleeding events by 20% compared to warfarin in patients with bioprosthetic mitral valves. In the ENAVLE trial, Edoxaban achieved a 3.7% decrease in thromboembolic events compared to warfarin without increasing significant bleeding rates. In the ENVISAGE-TAVI AF trial, edoxaban was noninferior to VKAs in preventing thromboembolic events but showed a slight increase in major bleeding events by 1.5%. Lastly, the ARCADIA trial highlighted that apixaban did not significantly reduce recurrent stroke risk compared to aspirin, with both treatments having an annualized stroke rate of 4.4%. Conclusion Advances in anticoagulant therapies and drug delivery systems aim to enhance patients' clinical outcomes for thromboembolic disorders. While recent trials show promising data, ongoing patient-specific responses and monitoring challenges require further research. Continuous innovation and investigation are essential to refine anticoagulation practices and tailor treatments.
Collapse
Affiliation(s)
- Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, Connecticut, USA
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, USA
| | - Abdur Jamil
- Department of Medicine, Samaritan Medical Centre Watertown, New York, USA
| | | | | |
Collapse
|
4
|
Chandiramani R, Mehta A, Blumenthal RS, Williams MS. Should We Use Aspirin or P2Y 12 Inhibitor Monotherapy in Stable Ischemic Heart Disease? Curr Atheroscler Rep 2024; 26:649-658. [PMID: 39243345 DOI: 10.1007/s11883-024-01234-2] [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] [Accepted: 08/22/2024] [Indexed: 09/09/2024]
Abstract
PURPOSE OF REVIEW To summarize the recent evidence and guideline recommendations on aspirin or P2Y12 inhibitor monotherapy in patients with stable ischemic heart disease and provide insights into future directions on this topic, which involves transition to a personalized assessment of bleeding and thrombotic risks. RECENT FINDINGS It has been questioned whether the evidence for aspirin as the foundational component of secondary prevention in patients with coronary artery disease aligns with contemporary pharmaco-invasive strategies. The recent HOST-EXAM study randomized patients who had received dual antiplatelet therapy for 6 to 18 months without ischemic or major bleeding events to either clopidogrel or aspirin for a further 24 months, and demonstrated that the patients in the clopidogrel arm had significantly lower rates of both thrombotic and bleeding complications compared to those in the aspirin arm. The patient-level PANTHER meta-analysis showed that in patients with established coronary artery disease, P2Y12 inhibitor monotherapy was associated with lower rates of myocardial infarction, stent thrombosis as well as gastrointestinal bleeding and hemorrhagic stroke compared to aspirin monotherapy, albeit with similar rates of all-cause mortality, cardiovascular mortality and major bleeding. Long-term low-dose aspirin is recommended for secondary prevention in patients with stable ischemic heart disease, with clopidogrel monotherapy being acknowledged as a feasible alternative. Dual antiplatelet therapy for six months after percutaneous coronary intervention remains the standard recommendation for patients with stable ischemic heart disease. However, the duration of dual antiplatelet therapy may be shortened and followed by P2Y12 inhibitor monotherapy or prolonged based on individualized evaluation of the patient's risk profile.
Collapse
Affiliation(s)
| | - Adhya Mehta
- Department of Internal Medicine, Albert Einstein College of Medicine/Jacobi Medical Center, Bronx, NY, USA
| | | | - Marlene S Williams
- Department of Medicine, Division of Cardiology, The Johns Hopkins University, 301 Mason Lord Drive, Suite 2400, Baltimore, MD, 21224, USA.
| |
Collapse
|
5
|
Wang L, Feng Z, Shen S, Wang S, Xing J, Huang R, Shen H, Yan P, Wang J, Zhang W, Liu Y, He W, Mo R. Stabilized Cell Membrane-Derived Vesicles by Lipid Anchoring for Enhanced Drug Delivery. ACS NANO 2024; 18:28081-28094. [PMID: 39360741 DOI: 10.1021/acsnano.4c07341] [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: 10/16/2024]
Abstract
A cell membrane-derived vesicle (MV) that has cell-mimicking features with characteristic functionalities holds vast appeal for biomimetic nanomedicine and drug delivery but suffers from a major limitation of innate fragility and poor stability. Herein, we report a lipid-anchoring strategy for stabilizing MV for enhanced drug delivery. An array of amphiphilic mono-acyl phosphatidylcholines (MPCs) with specific hydrophobic moieties are synthesized and readily engineered on MV based on their commendable aqueous solubility and efficient membrane insertability. Incorporation of MPCs containing rigid ring structures in the hydrophobic segment demonstrates the potency of stabilizing MV by the combined ordering and condensing effects. The optimized MPC-stabilized MV exhibits prolonged circulation in the bloodstream, elevated accumulation within a tumor, and enhanced therapeutic effects of chemotherapeutic and photothermal drugs. Moreover, doxorubicin-loaded MV, engineered with mono-all-trans retinoyl phosphatidylcholine as an MV stabilizer and a therapeutic prodrug, potently suppresses growth and metastasis of high-stemness tumors.
Collapse
Affiliation(s)
- Leikun Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Zhizi Feng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Shiyang Shen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Shengdi Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Jiaqi Xing
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Renqi Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - He Shen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Pengyi Yan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Jingyao Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Wenjing Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yiru Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Wei He
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Ran Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
6
|
Lv N, Zhai S, Xiong J, Hu N, Guo X, Liu Z, Zhang R. Enhanced-permeability delivery system for hydroxyl radical-responsive NIR-II fluorescence-monitored thrombolytic therapy. Colloids Surf B Biointerfaces 2024; 245:114193. [PMID: 39241635 DOI: 10.1016/j.colsurfb.2024.114193] [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: 05/15/2024] [Revised: 06/26/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024]
Abstract
Pathological thrombus can cause serious acute diseases that present a significant threat to human health, such as myocardial infarction and stroke. Challenges remain in achieving effective thrombolysis and real-time monitoring of therapeutic effects while minimizing side effects. Herein,a multifunctional nanoplatform (TG-OPDEA@UK/MnO2-H1080) with enhanced thrombus-permeability was developed to monitor the therapeutic effect of antioxidant-thrombolysis by hydroxyl radical-responsive NIR-II fluorescence imaging. The polyzwitterion poly (oxidized N,N-Diethylaminoethyl methacrylate-co-n-butyl methacrylate) (OPDEA) was prepared as the matrix of nanoparticles to simultaneously loading urokinase (UK) and MnO2 QDs, as well as NIR-II fluorescent molecule, H-1080. Subsequently, the fibrin targeted peptide CREKA was modified on the surface of the nanoparticles. OPDEA exhibits efficient loading capacity while endowing nanoparticles with the ability to effectively increased penetration depth of UK by 94.1 % into the thrombus, for extensive thrombolysis and fluorescence monitoring. The loaded UK exhibited good thrombolytic effect and greatly reduced the risk of bleeding by 82.6 %. TG-OPDEA@UK/MnO2-H1080 showed good thrombolytic efficacy and specific thrombus monitoring in the mouse carotid artery thrombosis model induced by ferric chloride (FeCl3). This work prepares a nanoplatform for thrombolytic therapy and real-time efficacy assessment based on an independent externally forced thrombus penetration delivery strategy.
Collapse
Affiliation(s)
- Nan Lv
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China
| | - Shaodong Zhai
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China; Shanxi Academy of Advanced Research and Innovation, Taiyuan 030032, China.
| | - Jun Xiong
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China
| | - Nan Hu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Xiang Guo
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Zhida Liu
- Shanxi Academy of Advanced Research and Innovation, Taiyuan 030032, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial Peoples Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
7
|
Puspitasari YM, Ministrini S, Han J, Karch C, Prisco F, Liberale L, Bengs S, Akhmedov A, Montecucco F, Beer JH, Lüscher TF, Bongiovanni D, Camici GG. Hutchinson-Gilford progeria syndrome mice display accelerated arterial thrombus formation and increased platelet reactivity. Thromb Res 2024; 241:109100. [PMID: 39032390 DOI: 10.1016/j.thromres.2024.109100] [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: 12/18/2023] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
INTRODUCTION Hutchinson-Gilford Progeria Syndrome (HGPS) is an ultra-rare premature aging genetic disorder caused by a point mutation in the lamin A gene, LMNA. Children with HGPS display short lifespans and typically die due to myocardial infarction or ischemic stroke, both acute cardiovascular events that are tightly linked to arterial thrombosis. Despite this fact, the effect of the classic HGPS LMNA gene mutation on arterial thrombosis remains unknown. METHODS Heterozygous LmnaG609G knock-in (LmnaG609G/+) mice, yielding an equivalent classic mutation observed in HGPS patients (c.1824C>T; pG608G mutation in the human LMNA gene) and corresponding wild-type (WT) control littermates underwent photochemically laser-induced carotid injury to trigger thrombosis. Coagulation and fibrinolytic factors were measured. Furthermore, platelet activation and reactivity were investigated. RESULTS LmnaG609G/+ mice displayed accelerated arterial thrombus formation, as underlined by shortened time to occlusion compared to WT littermates. Levels of factors involved in the coagulation and fibrinolytic system were comparable between groups, while LmnaG609G/+ animals showed higher plasma levels of thrombin-antithrombin complex and lower levels of antithrombin. Bone marrow analysis showed larger megakaryocytes in progeric mice. Lastly, enhanced platelet activation upon adenosine diphosphate, collagen-related peptide, and thrombin stimulation was observed in LmnaG609G/+ animals compared to the WT group, indicating a higher platelet reactivity in progeric animals. CONCLUSIONS LMNA mutation in HGPS mice accelerates arterial thrombus formation, which is mediated, at least in part, by enhanced platelet reactivity, which consequently augments thrombin generation. Given the wide spectrum of antiplatelet agents available clinically, further investigation is warranted to consider the most suitable antiplatelet regimen for children with HGPS to mitigate disease mortality and morbidity.
Collapse
Affiliation(s)
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Jiaying Han
- Department of Internal Medicine I, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Caroline Karch
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Francesco Prisco
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Susan Bengs
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Cardiology, Royal Brompton & Harefield Hospitals, National Heart & Lung Institute, Imperial College, London, United Kingdom
| | - Dario Bongiovanni
- Department of Internal Medicine I, Cardiology, University Hospital Augsburg, University of Augsburg, Augsburg, Germany; Department of Cardiovascular Medicine, Humanitas Clinical and Research Center IRCCS and Humanitas University, Rozzano, Milan, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Research and Education, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
8
|
Behrangzade A, Ye SH, Maestas DR, Wagner WR, Vande Geest JP. Improving the hemocompatibility of a porohyperelastic layered vascular graft using luminal reversal microflows. J Mech Behav Biomed Mater 2024; 157:106638. [PMID: 38996626 PMCID: PMC11513160 DOI: 10.1016/j.jmbbm.2024.106638] [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: 01/12/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 07/14/2024]
Abstract
Vascular graft thrombosis is a long-standing clinical problem. A myriad of efforts have been devoted to reducing thrombus formation following bypass surgery. Researchers have primarily taken a chemical approach to engineer and modify surfaces, seeking to make them more suitable for blood contacting applications. Using mechanical forces and surface topology to prevent thrombus formation has recently gained more attention. In this study, we have designed a bilayered porous vascular graft capable of repelling platelets and destabilizing absorbed protein layers from the luminal surface. During systole, fluid penetrates through the graft wall and is subsequently ejected from the wall into the luminal space (Luminal Reversal Flow - LRF), pushing platelets away from the surface during diastole. In-vitro hemocompatibility tests were conducted to compare platelet deposition in high LRF grafts with low LRF grafts. Graft material properties were determined and utilized in a porohyperelastic (PHE) finite element model to computationally predict the LRF generation in each graft type. Hemocompatibility testing showed significantly lower platelet deposition values in high versus low LRF generating grafts (median±IQR = 5,708 ± 987 and 23,039 ± 3,310 platelets per mm2, respectively, p=0.032). SEM imaging of the luminal surface of both graft types confirmed the quantitative blood test results. The computational simulations of high and low LRF generating grafts resulted in LRF values of -10.06 μm/s and -2.87 μm/s, respectively. These analyses show that a 250% increase in LRF is associated with a 75.2% decrease in platelet deposition. PHE vascular grafts with high LRF have the potential to improve anti-thrombogenicity and reduce thrombus-related post-procedure complications. Additional research is required to overcome the limitations of current graft fabrication technologies that further enhance LRF generation.
Collapse
Affiliation(s)
- Ali Behrangzade
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Sang-Ho Ye
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - David R Maestas
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - William R Wagner
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Jonathan P Vande Geest
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Mechanical Engineering and Material Science, University of Pittsburgh, Pittsburgh, PA, United States of America; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
9
|
Balmforth C, Whittington B, Tzolos E, Bing R, Williams MC, Clark L, Corral CA, Tavares A, Dweck MR, Newby DE. Translational molecular imaging: Thrombosis imaging with positron emission tomography. J Nucl Cardiol 2024; 39:101848. [PMID: 38499227 DOI: 10.1016/j.nuclcard.2024.101848] [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: 12/29/2023] [Revised: 03/05/2024] [Accepted: 03/10/2024] [Indexed: 03/20/2024]
Abstract
A key focus of cardiovascular medicine is the detection, treatment, and prevention of disease, with a move towards more personalized and patient-centred treatments. To achieve this goal, novel imaging approaches that allow for early and accurate detection of disease and risk stratification are needed. At present, the diagnosis, monitoring, and prognostication of thrombotic cardiovascular diseases are based on imaging techniques that measure changes in structural anatomy and biological function. Molecular imaging is emerging as a new tool for the non-invasive detection of biological processes, such as thrombosis, that can improve identification of these events above and beyond current imaging modalities. At the forefront of these evolving techniques is the use of high-sensitivity radiotracers in conjunction with positron emission tomography imaging that could revolutionise current diagnostic paradigms by improving our understanding of the role and origin of thrombosis in a range of cardiovascular diseases.
Collapse
Affiliation(s)
- Craig Balmforth
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom.
| | - Beth Whittington
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Evangelos Tzolos
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Rong Bing
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Michelle C Williams
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; Edinburgh Imaging, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Laura Clark
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Carlos Alcaide Corral
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; Edinburgh Imaging, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Adriana Tavares
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; Edinburgh Imaging, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Marc Richard Dweck
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - David Ernest Newby
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
10
|
Mehata AK, Bonlawar J, Tamang R, Malik AK, Setia A, Kumar S, Challa RR, Vallamkonda B, Koch B, Muthu MS. PLGA Nanoplatform for the Hypoxic Tumor Delivery: Folate Targeting, Therapy, and Ultrasound/Photoacoustic Imaging. ACS APPLIED BIO MATERIALS 2024; 7:5754-5770. [PMID: 39115968 DOI: 10.1021/acsabm.4c00853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Effective targeting of breast tumors is critical for improving therapeutic outcomes in breast cancer treatment. Additionally, hypoxic breast cancers are difficult to treat due to resistance toward chemotherapeutics, poor vascularity, and enhanced angiogenesis, which complicate effective drug delivery and therapeutic response. Addressing this formidable challenge requires designing a drug delivery system capable of targeted delivery of the anticancer agent, inhibition of efflux pump, and suppression of the tumor angiogenesis. Here, we have introduced Palbociclib (PCB)-loaded PLGA nanoparticles (NPs) consisting of chitosan-folate (CS-FOL) for folate receptor-targeted breast cancer therapy. The developed NPs were below 219 nm with a smooth, spherical surface shape. The entrapment efficiencies of NPs were achieved up to 85.78 ± 1.8%. Targeted NPs demonstrated faster drug release at pH 5.5, which potentiated the therapeutic efficacy of NPs due to the acidic microenvironment of breast cancer. In vitro cellular uptake study in MCF-7 cells confirmed the receptor-mediated endocytosis of targeted NPs. In vivo ultrasound and photoacoustic imaging studies on rats with hypoxic breast cancer showed that targeted NPs significantly reduced tumor growth and hypoxic tumor volume, and suppressed angiogenesis.
Collapse
Affiliation(s)
- Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| | - Jyoti Bonlawar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| | - Rupen Tamang
- Genotoxicology and Cancer Biology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Ankit Kumar Malik
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| | - Aseem Setia
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| | - Shailendra Kumar
- SATHI, Central Discovery Centre, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Ranadheer Reddy Challa
- Department of Pharmaceutical Science, School of Applied Sciences and Humanities, VIGNAN's Foundation for Science, Technology & Research, Vadlamudi 522213, Andhra Pradesh, India
| | - Bhaskar Vallamkonda
- Department of Pharmaceutical Science, School of Applied Sciences and Humanities, VIGNAN's Foundation for Science, Technology & Research, Vadlamudi 522213, Andhra Pradesh, India
| | - Biplob Koch
- Genotoxicology and Cancer Biology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| |
Collapse
|
11
|
Huang Y, Wang J, Guo Y, Shen L, Li Y. Fibrinogen binding to activated platelets and its biomimetic thrombus-targeted thrombolytic strategies. Int J Biol Macromol 2024; 274:133286. [PMID: 38908635 DOI: 10.1016/j.ijbiomac.2024.133286] [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: 11/24/2023] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
Thrombosis is associated with various fatal arteriovenous syndromes including ischemic stroke, myocardial infarction, and pulmonary embolism. However, current clinical thrombolytic treatment strategies still have many problems in targeting and safety to meet the thrombolytic therapy needs. Understanding the molecular mechanism that underlies thrombosis is critical in developing effective thrombolytic strategies. It is well known that platelets play a central role in thrombosis and the binding of fibrinogen to activated platelets is a common pathway in the process of clot formation. Based on this, a concept of biomimetic thrombus-targeted thrombolytic strategy inspired from fibrinogen binding to activated platelets in thrombosis was proposed, which could selectively bind to activated platelets at a thrombus site, thus enabling targeted delivery and local release of thrombolytic agents for effective thrombolysis. In this review, we first summarized the main characteristics of platelets and fibrinogen, and then introduced the classical molecular mechanisms of thrombosis, including platelet adhesion, platelet activation and platelet aggregation through the interactions of activated platelets with fibrinogen. In addition, we highlighted the recent advances in biomimetic thrombus-targeted thrombolytic strategies which inspired from fibrinogen binding to activated platelets in thrombosis. The possible future directions and perspectives in this emerging area are briefly discussed.
Collapse
Affiliation(s)
- Yu Huang
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China.
| | - Jiahua Wang
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China
| | - Yuanyuan Guo
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China
| | - Lingyue Shen
- Department of Oral & Maxillofacial-Head & Neck Oncology, Department of Laser and Aesthetic Medicine, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stoma-tology & Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai 200011, PR China.
| | - Yuehua Li
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China.
| |
Collapse
|
12
|
Vandelanotte S, De Meyer SF. Acute Ischemic Stroke Thrombus Composition. Neuroscience 2024; 550:11-20. [PMID: 38185279 DOI: 10.1016/j.neuroscience.2023.12.010] [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: 10/27/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/09/2024]
Abstract
Ischemic stroke is caused by a thrombus blocking one or multiple arteries in the brain, resulting in irreversible damage in the associated brain tissue. The aim of therapy is to restore the blood flow as fast as possible. Two recanalization strategies are currently available: pharmacological thrombolysis using recombinant tissue plasminogen activator (rt-PA) and mechanical removal of the thrombus. Despite recent advancements, achieving efficient recanalization remains a challenge. The precise causes of therapy failure are not fully understood but thrombus composition is likely a key factor in successful recanalization. This review explores acute ischemic stroke thrombus composition, its recently identified components, and how it affects stroke treatment. It also discusses how new insights could enhance current recanalization strategies for ischemic stroke patients.
Collapse
Affiliation(s)
| | - Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium.
| |
Collapse
|
13
|
Liao W, Lu Z, Wang C, Zhu X, Yang Y, Zhou Y, Gong P. Application and advances of biomimetic membrane materials in central nervous system disorders. J Nanobiotechnology 2024; 22:280. [PMID: 38783302 PMCID: PMC11112845 DOI: 10.1186/s12951-024-02548-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Central nervous system (CNS) diseases encompass spinal cord injuries, brain tumors, neurodegenerative diseases, and ischemic strokes. Recently, there has been a growing global recognition of CNS disorders as a leading cause of disability and death in humans and the second most common cause of death worldwide. The global burdens and treatment challenges posed by CNS disorders are particularly significant in the context of a rapidly expanding global population and aging demographics. The blood-brain barrier (BBB) presents a challenge for effective drug delivery in CNS disorders, as conventional drugs often have limited penetration into the brain. Advances in biomimetic membrane nanomaterials technology have shown promise in enhancing drug delivery for various CNS disorders, leveraging properties such as natural biological surfaces, high biocompatibility and biosafety. This review discusses recent developments in biomimetic membrane materials, summarizes the types and preparation methods of these materials, analyzes their applications in treating CNS injuries, and provides insights into the future prospects and limitations of biomimetic membrane materials.
Collapse
Affiliation(s)
- Weiquan Liao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Zhichao Lu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Chenxing Wang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Xingjia Zhu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Yang Yang
- Department of Trauma Center, Affiliated Hospital of Nantong University, Medical school of Nantong University, Nantong, Jiangsu, 226001, China
| | - Youlang Zhou
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.
| | - Peipei Gong
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, 226001, China.
- Jiangsu Medical Innovation Center, Neurological Disease Diagnosis and Treatment Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
14
|
Nguyen HT, Vu MP, Nguyen TTM, Nguyen TT, Kieu TVO, Duong HY, Pham PT, Hoang TH. Association of the neutrophil-to-lymphocyte ratio with the occurrence of venous thromboembolism and arterial thrombosis. J Int Med Res 2024; 52:3000605241240999. [PMID: 38606734 PMCID: PMC11015807 DOI: 10.1177/03000605241240999] [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: 11/29/2023] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
OBJECTIVE This study aimed to assess the association of the neutrophil-to-lymphocyte ratio (NLR) with the occurrence of venous thromboembolism (VTE) and arterial thrombosis (AT). METHODS This was a retrospective cross-sectional study including 585 medical records obtained from all consecutive patients who were suspected of having thrombosis. RESULTS The AT group had a higher neutrophil count and NLR and a lower lymphocyte count than the non-thrombosis group. Receiver operating characteristic curve analysis showed the ability of the NLR to predict the presence of AT. The cut-off value for the NLR was 4.44. No distinction was found in the NLR between the VTE and non-thrombosis groups. Regression analysis showed that a high NLR was an independent factor related to the presence of AT. Patients with an NLR ≥ 4.44 had a higher risk of AT than those with an NLR < 4.44 (odds ratio = 2.015, 95% confidence interval: 1.180-3.443). CONCLUSION A high NLR may be considered a predictive factor for the occurrence of AT, but an association with the presence of VTE was not found.
Collapse
Affiliation(s)
- Ha Thanh Nguyen
- Department of Hematology, Hanoi Medical University, Hanoi, Vietnam
| | - Minh Phuong Vu
- Department of Hematology, Hanoi Medical University, Hanoi, Vietnam
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| | - Thi Tuyet Mai Nguyen
- Department of Hematology, Hanoi Medical University, Hanoi, Vietnam
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| | - Tuan Tung Nguyen
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| | - Thi Van Oanh Kieu
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| | - Hai Yen Duong
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| | - Phuong Thao Pham
- Department of Hematology, Hanoi Medical University, Hanoi, Vietnam
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| | - Thi Hue Hoang
- Department of Hematology, Hanoi Medical University, Hanoi, Vietnam
- Hematology and Blood Transfusion Center, Bach Mai Hospital, Hanoi, Vietnam
| |
Collapse
|
15
|
Spirito A, Krishnan SL, Capodanno D, Angiolillo DJ, Mehran R. Antiplatelet De-Escalation Strategies in Patients Undergoing Percutaneous Coronary Intervention. Circ Cardiovasc Interv 2024; 17:e013263. [PMID: 38626078 DOI: 10.1161/circinterventions.123.013263] [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] [Indexed: 04/18/2024]
Abstract
Dual antiplatelet therapy-the combination of aspirin and a P2Y12 inhibitor-remains the standard antiplatelet regimen recommended to prevent ischemic complications immediately after percutaneous coronary intervention. Nonetheless, recent advances in stent technologies, percutaneous coronary intervention techniques, adjunctive pharmacotherapy for secondary prevention, and the rising awareness of the prognostic impact of bleeding, which are inevitably associated with dual antiplatelet therapy, led to the investigation of alternative antiplatelet regimens related to fewer bleeding and a preserved ischemic protection. Thrombotic complications occur mostly in the first months after percutaneous coronary intervention, while the risk of bleeding remains stable over time; this observation laid the foundation of the concept of antiplatelet de-escalation, consisting of a more intense antiplatelet regimen early after percutaneous coronary intervention, followed by a less potent antiplatelet therapy thereafter. According to new definitions proposed by the Academic Research Consortium, de-escalation can be achieved by discontinuation of 1 antiplatelet agent, switching from a potent P2Y12 inhibitor to clopidogrel, or by reducing the dose of antiplatelet agents. This review discusses the rationale and the evidence supporting antiplatelet de-escalation, provides practical guidance to use these new regimens, and gives insights into future developments in the field.
Collapse
Affiliation(s)
- Alessandro Spirito
- The Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY (A.S., S.L.K., R.M.)
| | - Sriya L Krishnan
- The Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY (A.S., S.L.K., R.M.)
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero Universitaria Policlinico "G. Rodolico-San Marco," Catania, Italy (D.C.)
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville (D.J.A.)
| | - Roxana Mehran
- The Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY (A.S., S.L.K., R.M.)
| |
Collapse
|
16
|
Ishida M, Sakai C, Kobayashi Y, Ishida T. Cigarette Smoking and Atherosclerotic Cardiovascular Disease. J Atheroscler Thromb 2024; 31:189-200. [PMID: 38220184 PMCID: PMC10918046 DOI: 10.5551/jat.rv22015] [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: 10/31/2023] [Accepted: 12/01/2023] [Indexed: 01/16/2024] Open
Abstract
The detrimental effects of cigarette smoking on cardiovascular health, particularly atherosclerosis and thrombosis, are well established, and more detailed mechanisms continue to emerge. As the fundamental pathophysiology of the adverse effects of smoking, endothelial dysfunction, inflammation, and thrombosis are considered to be particularly important. Cigarette smoke induces endothelial dysfunction, leading to impaired vascular dilation and hemostasis regulation. Factors contributing to endothelial dysfunction include reduced bioavailability of nitric oxide, increased levels of superoxide anion, and endothelin release. Chronic inflammation of the vascular wall is a central pathogenesis of smoking-induced atherosclerosis. Smoking systemically elevates inflammatory markers and induces the expression of adhesion molecules and cytokines in various tissues. Pattern recognition receptors and damage-associated molecular patterns play crucial roles in the mechanism underlying smoking-induced inflammation. Smoking-induced DNA damage and activation of innate immunity, such as the NLRP3 inflammasome, cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, and Toll-like receptor 9, are shown to amplify inflammatory cytokine expression. Cigarette smoke-induced oxidative stress and inflammation influence platelet adhesion, aggregation, and coagulation via adhesion molecule upregulation. Furthermore, it affects the coagulation cascade and fibrinolysis balance, causing thrombus formation. Matrix metalloproteinases contribute to plaque vulnerability and atherothrombotic events. The impact of smoking on inflammatory cells and adhesion molecules further intensifies the risk of atherothrombosis. Collectively, exposure to cigarette smoke exerts profound effects on endothelial function, inflammation, and thrombosis, contributing to the development and progression of atherosclerosis and atherothrombotic cardiovascular diseases. Understanding these intricate mechanisms highlights the urgent need for smoking cessation to protect cardiovascular health. This comprehensive review investigates the multifaceted mechanisms through which smoking contributes to these life-threatening conditions.
Collapse
Affiliation(s)
- Mari Ishida
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Chiemi Sakai
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yusuke Kobayashi
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takafumi Ishida
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
17
|
Liu X, Wang L, Xiang Y, Liao F, Li N, Li J, Wang J, Wu Q, Zhou C, Yang Y, Kou Y, Yang Y, Tang H, Zhou N, Wan C, Yin Z, Yang GZ, Tao G, Zang J. Magnetic soft microfiberbots for robotic embolization. Sci Robot 2024; 9:eadh2479. [PMID: 38381840 DOI: 10.1126/scirobotics.adh2479] [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: 02/19/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024]
Abstract
Cerebral aneurysms and brain tumors are leading life-threatening diseases worldwide. By deliberately occluding the target lesion to reduce the blood supply, embolization has been widely used clinically to treat cerebral aneurysms and brain tumors. Conventional embolization is usually performed by threading a catheter through blood vessels to the target lesion, which is often limited by the poor steerability of the catheter in complex neurovascular networks, especially in submillimeter regions. Here, we propose magnetic soft microfiberbots with high steerability, reliable maneuverability, and multimodal shape reconfigurability to perform robotic embolization in submillimeter regions via a remote, untethered, and magnetically controllable manner. Magnetic soft microfiberbots were fabricated by thermal drawing magnetic soft composite into microfibers, followed by magnetizing and molding procedures to endow a helical magnetic polarity. By controlling magnetic fields, magnetic soft microfiberbots exhibit reversible elongated/aggregated shape morphing and helical propulsion in flow conditions, allowing for controllable navigation through complex vasculature and robotic embolization in submillimeter regions. We performed in vitro embolization of aneurysm and tumor in neurovascular phantoms and in vivo embolization of a rabbit femoral artery model under real-time fluoroscopy. These studies demonstrate the potential clinical value of our work, paving the way for a robotic embolization scheme in robotic settings.
Collapse
Affiliation(s)
- Xurui Liu
- School of Integrated Circuits, Huazhong University of Science and Technology, Wuhan 430074, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Liu Wang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei 230026, PR China
- State Key Laboratory of Nonlinear Mechanics, Institute of Mechanics, Chinese Academy of Science, 15 Beisihuan West Road, Beijing 100190, China
| | - Yuanzhuo Xiang
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Fan Liao
- School of Integrated Circuits, Huazhong University of Science and Technology, Wuhan 430074, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Na Li
- School of Integrated Circuits, Huazhong University of Science and Technology, Wuhan 430074, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiyu Li
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei 230026, PR China
| | - Jiaxin Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Qingyang Wu
- School of Integrated Circuits, Huazhong University of Science and Technology, Wuhan 430074, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Cheng Zhou
- School of Integrated Circuits, Huazhong University of Science and Technology, Wuhan 430074, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Youzhou Yang
- School of Integrated Circuits, Huazhong University of Science and Technology, Wuhan 430074, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yuanshi Kou
- School of Integrated Circuits, Huazhong University of Science and Technology, Wuhan 430074, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yueying Yang
- School of Integrated Circuits, Huazhong University of Science and Technology, Wuhan 430074, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hanchuan Tang
- School of Integrated Circuits, Huazhong University of Science and Technology, Wuhan 430074, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ning Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430074, China
| | - Chidan Wan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhouping Yin
- Flexible Electronics Research Center, State Key Laboratory of Digital Manufacturing Equipment and Technology, School of Mechanical Science and Engineering, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- State Key Laboratory of Intelligent Manufacturing Equipment and Technology, School of Mechanical Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Guang-Zhong Yang
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guangming Tao
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
- State Key Laboratory of Material Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Institute of Medical Equipment Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jianfeng Zang
- School of Integrated Circuits, Huazhong University of Science and Technology, Wuhan 430074, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
- State Key Laboratory of Intelligent Manufacturing Equipment and Technology, School of Mechanical Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
18
|
He X, Dutta S, Liang J, Paul C, Huang W, Xu M, Chang V, Ao I, Wang Y. Direct cellular reprogramming techniques for cardiovascular regenerative therapeutics. Can J Physiol Pharmacol 2024; 102:1-13. [PMID: 37903419 DOI: 10.1139/cjpp-2023-0088] [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] [Indexed: 11/01/2023]
Abstract
Cardiovascular diseases remain a leading cause of hospitalization affecting approximately 38 million people worldwide. While pharmacological and revascularization techniques can improve the patient's survival and quality of life, they cannot help reversing myocardial infarction injury and heart failure. Direct reprogramming of somatic cells to cardiomyocyte and cardiac progenitor cells offers a new approach to cellular reprogramming and paves the way for translational regenerative medicine. Direct reprogramming can bypass the pluripotent stage with the potential advantage of non-immunogenic cell products, reduced carcinogenic risk, and no requirement for embryonic tissue. The process of directly reprogramming cardiac cells was first achieved through the overexpression of transcription factors such as GATA4, MEF2C, and TBX5. However, over the past decade, significant work has been focused on enhancing direct reprogramming using a mixture of transcription factors, microRNAs, and small molecules to achieve cardiac cell fate. This review discusses the evolution of direct reprogramming, recent progress in achieving efficient cardiac cell fate conversion, and describes the reprogramming mechanisms at a molecular level. We also explore various viral and non-viral delivery methods currently being used to aid in the delivery of reprogramming factors to improve efficiency. However, further studies will be needed to overcome molecular and epigenetic barriers to successfully achieve translational cardiac regenerative therapeutics.
Collapse
Affiliation(s)
- Xingyu He
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Suchandrima Dutta
- Department of Internal MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Jialiang Liang
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Christian Paul
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Wei Huang
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Meifeng Xu
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Vivian Chang
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Ian Ao
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Yigang Wang
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| |
Collapse
|
19
|
Zhong T, Gao N, Guan Y, Liu Z, Guan J. Co-Delivery of Bioengineered Exosomes and Oxygen for Treating Critical Limb Ischemia in Diabetic Mice. ACS NANO 2023; 17:25157-25174. [PMID: 38063490 PMCID: PMC10790628 DOI: 10.1021/acsnano.3c08088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Diabetic patients with critical limb ischemia face a high rate of limb amputation. Regeneration of the vasculature and skeletal muscles can salvage diseased limbs. Therapy using stem cell-derived exosomes that contain multiple proangiogenic and promyogenic factors represents a promising strategy. Yet the therapeutic efficacy is not optimal because exosomes alone cannot efficiently rescue and recruit endothelial and skeletal muscle cells and restore their functions under hyperglycemic and ischemic conditions. To address these limitations, we fabricated ischemic-limb-targeting stem cell-derived exosomes and oxygen-releasing nanoparticles and codelivered them in order to recruit endothelial and skeletal muscle cells, improve cell survival under ischemia before vasculature is established, and restore cell morphogenic function under high glucose and ischemic conditions. The exosomes and oxygen-releasing nanoparticles, delivered by intravenous injection, specifically accumulated in the ischemic limbs. Following 4 weeks of delivery, the exosomes and released oxygen synergistically stimulated angiogenesis and muscle regeneration without inducing substantial inflammation and reactive oxygen species overproduction. Our work demonstrates that codelivery of exosomes and oxygen is a promising treatment solution for saving diabetic ischemic limbs.
Collapse
Affiliation(s)
- Ting Zhong
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Ning Gao
- Institute of Materials Science and Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Ya Guan
- Institute of Materials Science and Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Zhongting Liu
- Institute of Materials Science and Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Jianjun Guan
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Institute of Materials Science and Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| |
Collapse
|
20
|
Fischbach A, Lamberti M, Simons JA, Wrede E, Theißen A, Winnersbach P, Rossaint R, Stollenwerk A, Bleilevens C. Early Blood Clot Detection Using Forward Scattering Light Measurements Is Not Superior to Delta Pressure Measurements. BIOSENSORS 2023; 13:1012. [PMID: 38131772 PMCID: PMC10741584 DOI: 10.3390/bios13121012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023]
Abstract
The occurrence of thrombus formation within an extracorporeal membrane oxygenator is a common complication during extracorporeal membrane oxygenation therapy and can rapidly result in a life-threatening situation due to arterial thromboembolism, causing stroke, pulmonary embolism, and limb ischemia in the patient. The standard clinical practice is to monitor the pressure at the inlet and outlet of oxygenators, indicating fulminant, obstructive clot formation indicated by an increasing pressure difference (ΔP). However, smaller blood clots at early stages are not detectable. Therefore, there is an unmet need for sensors that can detect blood clots at an early stage to minimize the associated thromboembolic risks for patients. This study aimed to evaluate if forward scattered light (FSL) measurements can be used for early blood clot detection and if it is superior to the current clinical gold standard (pressure measurements). A miniaturized in vitro test circuit, including a custom-made test chamber, was used. Heparinized human whole blood was circulated through the test circuit until clot formation occurred. Four LEDs and four photodiodes were placed along the sidewall of the test chamber in different positions for FSL measurements. The pressure monitor was connected to the inlet and the outlet to detect changes in ΔP across the test chamber. Despite several modifications in the LED positions on the test chamber, the FSL measurements could not reliably detect a blood clot within the in vitro test circuit, although the pressure measurements used as the current clinical gold standard detected fulminant clot formation in 11 independent experiments.
Collapse
Affiliation(s)
- Anna Fischbach
- Department of Anesthesiology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.A.S.); (A.T.); (P.W.); (R.R.)
| | - Michael Lamberti
- Informatics 11—Embedded Software, RWTH Aachen University, 52074 Aachen, Germany; (M.L.); (E.W.); (A.S.)
| | - Julia Alexandra Simons
- Department of Anesthesiology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.A.S.); (A.T.); (P.W.); (R.R.)
| | - Erik Wrede
- Informatics 11—Embedded Software, RWTH Aachen University, 52074 Aachen, Germany; (M.L.); (E.W.); (A.S.)
| | - Alexander Theißen
- Department of Anesthesiology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.A.S.); (A.T.); (P.W.); (R.R.)
| | - Patrick Winnersbach
- Department of Anesthesiology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.A.S.); (A.T.); (P.W.); (R.R.)
| | - Rolf Rossaint
- Department of Anesthesiology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.A.S.); (A.T.); (P.W.); (R.R.)
| | - André Stollenwerk
- Informatics 11—Embedded Software, RWTH Aachen University, 52074 Aachen, Germany; (M.L.); (E.W.); (A.S.)
| | - Christian Bleilevens
- Department of Anesthesiology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.A.S.); (A.T.); (P.W.); (R.R.)
| |
Collapse
|
21
|
Ince O, Gulsen K, Ozcan S, Donmez E, Ziyrek M, Sahin I, Okuyan E. Is dynamic change in mean platelet volume related with composite endpoint development after transcatheter aortic valve replacement? Blood Coagul Fibrinolysis 2023; 34:487-493. [PMID: 37756207 DOI: 10.1097/mbc.0000000000001255] [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: 09/29/2023]
Abstract
Aortic valve stenosis (AS) is the most common valvular disease, and surgical or transcatheter aortic valve replacement (TAVR) are the treatment options. Diminish in platelet production or dysfunction may occur due to shear stress, advanced age, and other coexisting diseases in AS patients. Bleeding is one of the complications of TAVR and associated with increased mortality. MPV (mean platelet volume) indicates platelet's thrombogenic activity. Overproduction or consumption of platelets in various cardiac conditions may affect MPV values. We aimed to investigate the pre and postprocedure MPV percentage change (MPV-PC) and its association with post-TAVR short-term complications. A total of 204 patients who underwent TAVR with a diagnosis of severe symptomatic AS were included. The mean age was 78.66 ± 6.45 years, and 49.5% of patients were women. Two groups generated according to composite end point (CEP) development: CEP(+) and CEP(-).110 patients(53.9%) formed CEP(+) group. Although baseline MPV and platelet levels were similar between groups, MPV was increased ( P < 0.001) and platelet was decreased ( P < 0.001) significantly following the procedure when compared to baseline. MPV-PC was significantly higher in the VARC type 2-4 bleeding ( P = 0.036) and major vascular, access-related, or cardiac structural complication groups ( P = 0.048) when CEP subgroups were analyzed individually. Regression analysis revealed that diabetes mellitus [ P = 0.044, β: 1.806 odds ratio (95% confidence interval): 1.016-3.21] and MPV-PC [ P = 0.007,β: 1.044 odds ratio (95% confidence interval): 1.012-1.077] as independent predictors of CEP development at 1 month after TAVR. The MPV increase following TAVR may be an indicator of adverse outcomes following TAVR procedure within 1-month.
Collapse
Affiliation(s)
- Orhan Ince
- Department of Cardiology, Istanbul Bagcilar Training and Research Hospital
| | - Kamil Gulsen
- Department of Cardiology, Health and Science University Kartal Kosuyolu Training and Research Hospital, Istanbul
| | - Sevgi Ozcan
- Department of Cardiology, Istanbul Bagcilar Training and Research Hospital
| | - Esra Donmez
- Department of Cardiology, Istanbul Bagcilar Training and Research Hospital
| | - Murat Ziyrek
- Department of Cardiology, Konya Farabi Hospital, Konya, Turkey
| | - Irfan Sahin
- Department of Cardiology, Istanbul Bagcilar Training and Research Hospital
| | - Ertugrul Okuyan
- Department of Cardiology, Istanbul Bagcilar Training and Research Hospital
| |
Collapse
|
22
|
Zhang X, Zhang Y, Liu F, Zhu J, Liang X, Shi X, Han L, Xu K, Cheng H. Red blood cell distribution width as a prognostic factor in patients with aplastic anemia treated with cyclosporin A plus androgen or cyclosporine A alone: a retrospective study. Hematology 2023; 28:2240665. [PMID: 37594305 DOI: 10.1080/16078454.2023.2240665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/20/2023] [Indexed: 08/19/2023] Open
Abstract
OBJECTIVE To explore the prognostic value of red blood cell distribution width (RDW) in newly diagnosed aplastic anemia (AA) patients treated with cyclosporine A (CsA) plus androgen or CsA alone. METHODS We retrospectively analyzed the clinical outcome of 220 patients with AA. According to the baseline level of RDW before treatment, the patients were divided into the high-RDW group (RDW ≥ 15%) and the normal-RDW group (RDW < 15%). RESULTS The median RDW of non-severe AA (NSAA) and severe AA (SAA) patients was 15.65% and 15.35%, respectively; this were significantly higher than that of very severe AA (VSAA) patients (13.35%). With median follow-up being 46 months, AA patients in the high-RDW group showed better 5-year OS and PFS than the normal-RDW group (93%: 75.3%; 74.3%: 61%). There was a higher ORR in the high-RDW group than the normal-RDW group (68.7%: 52.3%). The ORR of NSAA patients in the high-RDW group was better than that in the normal RDW group (75.8%: 60%). The 5-year OS of SAA/VSAA patients in the high-RDW group was significantly higher than the normal-RDW group (81.8%: 50.8%). CONCLUSION This is the first documentation on the prognostic value of RDW in AA patients receiving CsA treatment with long-term follow-up, which had shown that high RDW at diagnosis was a better prognostic factor.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Yanan Zhang
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Fengan Liu
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Jingjing Zhu
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Xiuli Liang
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Xuedong Shi
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Li Han
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Kailin Xu
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
- Institute of Hematology, Xuzhou Medical University, Jiangsu, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, People's Republic of China
| | - Hai Cheng
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| |
Collapse
|
23
|
Burnouf T, Chou ML, Lundy DJ, Chuang EY, Tseng CL, Goubran H. Expanding applications of allogeneic platelets, platelet lysates, and platelet extracellular vesicles in cell therapy, regenerative medicine, and targeted drug delivery. J Biomed Sci 2023; 30:79. [PMID: 37704991 PMCID: PMC10500824 DOI: 10.1186/s12929-023-00972-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023] Open
Abstract
Platelets are small anucleated blood cells primarily known for their vital hemostatic role. Allogeneic platelet concentrates (PCs) collected from healthy donors are an essential cellular product transfused by hospitals to control or prevent bleeding in patients affected by thrombocytopenia or platelet dysfunctions. Platelets fulfill additional essential functions in innate and adaptive immunity and inflammation, as well as in wound-healing and tissue-repair mechanisms. Platelets contain mitochondria, lysosomes, dense granules, and alpha-granules, which collectively are a remarkable reservoir of multiple trophic factors, enzymes, and signaling molecules. In addition, platelets are prone to release in the blood circulation a unique set of extracellular vesicles (p-EVs), which carry a rich biomolecular cargo influential in cell-cell communications. The exceptional functional roles played by platelets and p-EVs explain the recent interest in exploring the use of allogeneic PCs as source material to develop new biotherapies that could address needs in cell therapy, regenerative medicine, and targeted drug delivery. Pooled human platelet lysates (HPLs) can be produced from allogeneic PCs that have reached their expiration date and are no longer suitable for transfusion but remain valuable source materials for other applications. These HPLs can substitute for fetal bovine serum as a clinical grade xeno-free supplement of growth media used in the in vitro expansion of human cells for transplantation purposes. The use of expired allogeneic platelet concentrates has opened the way for small-pool or large-pool allogeneic HPLs and HPL-derived p-EVs as biotherapy for ocular surface disorders, wound care and, potentially, neurodegenerative diseases, osteoarthritis, and others. Additionally, allogeneic platelets are now seen as a readily available source of cells and EVs that can be exploited for targeted drug delivery vehicles. This article aims to offer an in-depth update on emerging translational applications of allogeneic platelet biotherapies while also highlighting their advantages and limitations as a clinical modality in regenerative medicine and cell therapies.
Collapse
Affiliation(s)
- Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan.
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Ming-Li Chou
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - David J Lundy
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Ching-Li Tseng
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Hadi Goubran
- Saskatoon Cancer Centre and College of Medicine, University of Saskatchewan, Saskatchewan, Canada
| |
Collapse
|
24
|
Huang S, Rodriguez C, Shakfeh K, Smith J, Reddy K. Urgent Revascularization of the Left Proximal Circumflex Following Cessation of Cangrelor Within Six Hours of Procedure. Cureus 2023; 15:e40314. [PMID: 37448385 PMCID: PMC10337834 DOI: 10.7759/cureus.40314] [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: 04/28/2023] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
Patients undergoing procedures are often transitioned off anticoagulants using anti-platelet agents with short half-lives as a "bridge." We present the case of a patient with a history of in-stent thromboses who experienced a thrombotic event following a literature-guided bridging protocol. This case is one of the first to show that stopping cangrelor within six hours led to a need for urgent revascularization and suggests that the timing for discontinuing bridging agents should be customized based on the patient's history of increased blood clotting.
Collapse
Affiliation(s)
- Sherri Huang
- Internal Medicine and Pediatrics, University of South Florida Morsani College of Medicine, Tampa, USA
| | - Camilo Rodriguez
- Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, USA
| | - Khalid Shakfeh
- Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, USA
| | - Jorden Smith
- Internal Medicine, University of South Florida Morsani College, Tampa, USA
| | - Koushik Reddy
- Cardiology and Lifestyle Medicine, James A. Haley Veterans Affair Medical Center, Tampa, USA
| |
Collapse
|
25
|
Heo J, Park JH, Kim HJ, Pahk K, Pahk KJ. Sonothrombolysis with an acoustic net-assisted boiling histotripsy: A proof-of-concept study. ULTRASONICS SONOCHEMISTRY 2023; 96:106435. [PMID: 37178667 DOI: 10.1016/j.ultsonch.2023.106435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023]
Abstract
Whilst sonothrombolysis is a promising and noninvasive ultrasound technique for treating blood clots, bleeding caused by thrombolytic agents used for dissolving clots and potential obstruction of blood flow by detached clots (i.e., embolus) are the major limitations of the current approach. In the present study, a new sonothrombolysis method is proposed for treating embolus without the use of thrombolytic drugs. Our proposed method involves (a) generating a spatially localised acoustic radiation force in a blood vessel against the blood flow to trap moving blood clots (i.e., generation of an acoustic net), (b) producing acoustic cavitation to mechanically destroy the trapped embolus, and (c) acoustically monitoring the trapping and mechanical fractionation processes. Three different ultrasound transducers with different purposes were employed in the proposed method: (1) 1-MHz dual focused ultrasound (dFUS) transducers for capturing moving blood clots, (2) a 2-MHz High Intensity Focused Ultrasound (HIFU) source for fractionating blood clots and (3) a passive acoustic emission detector with broad bandwidth (10 kHz to 20 MHz) for receiving and analysing acoustic waves scattered from a trapped embolus and acoustic cavitation. To demonstrate the feasibility of the proposed method, in vitro experiments with an optically transparent blood vessel-mimicking phantom filled with a blood mimicking fluid and a blood clot (1.2 to 5 mm in diameter) were performed with varying the dFUS and HIFU exposure conditions under various flow conditions (from 1.77 to 6.19 cm/s). A high-speed camera was used to observe the production of acoustic fields, acoustic cavitation formation and blood clot fragmentation within a blood vessel by the proposed method. Numerical simulations of acoustic and temperature fields generated under a given exposure condition were also conducted to further interpret experimental results on the proposed sonothrombolysis. Our results clearly showed that fringe pattern-like acoustic pressure fields (fringe width of 1 mm) produced in a blood vessel by the dFUS captured an embolus (1.2 to 5 mm in diameter) at the flow velocity up to 6.19 cm/s. This was likely to be due to the greater magnitude of the dFUS-induced acoustic radiation force exerted on an embolus in the opposite direction to the flow in a blood vessel than that of the drag force produced by the flow. The acoustically trapped embolus was then mechanically destructed into small pieces of debris (18 to 60 μm sized residual fragments) by the HIFU-induced strong cavitation without damaging the blood vessel walls. We also observed that acoustic emissions emitted from a blood clot captured by the dFUS and cavitation produced by the HIFU were clearly distinguished in the frequency domain. Taken together, these results can suggest that our proposed sonothrombolysis method could be used as a promising tool for treating thrombosis and embolism through capturing and destroying blood clots effectively.
Collapse
Affiliation(s)
- Jeongmin Heo
- Bionics Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Jun Hong Park
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Hyo Jun Kim
- LAAS-CNRS, University of Toulouse, CNRS, Toulouse, France
| | - Kisoo Pahk
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Ki Joo Pahk
- Department of Biomedical Engineering, Kyung Hee University, Yongin 17104, Republic of Korea.
| |
Collapse
|
26
|
Zhang Y, Liu Y, Ni G, Xu J, Tian Y, Liu X, Gao J, Gao Q, Shen Y, Yan Z. Sulfated modification, basic characterization, antioxidant and anticoagulant potentials of polysaccharide from Sagittaria trifolia. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
|
27
|
Gauer JS, Duval C, Xu RG, Macrae FL, McPherson HR, Tiede C, Tomlinson D, Watson SP, Ariëns RAS. Fibrin-glycoprotein VI interaction increases platelet procoagulant activity and impacts clot structure. J Thromb Haemost 2023; 21:667-681. [PMID: 36696196 DOI: 10.1016/j.jtha.2022.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/14/2022] [Accepted: 09/28/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND The glycoprotein VI (GPVI) signaling pathway was previously reported to direct procoagulant platelet activity through collagen binding. However, the impact of GPVI-fibrin interaction on procoagulant platelet development and how it modulates the clot structure are unknown. OBJECTIVES To determine the effect of GPVI-fibrin interaction on the platelet phenotype and its impact on the clot structure. METHODS Procoagulant platelets in platelet-rich plasma clots were determined by scanning electron microscopy (wild-type and GPVI-deficient murine samples) and confocal microscopy. Procoagulant platelet number, clot density, clot porosity, and clot retraction were determined in platelet-rich plasma or whole blood clots of healthy volunteers in the presence of tyrosine kinase inhibitors (PRT-060318, ibrutinib, and dasatinib) and eptifibatide. RESULTS GPVI-deficient clots showed a higher nonprocoagulant vs procoagulant platelet ratio than wild-type clots. The fiber density and the procoagulant platelet number decreased in the presence of Affimer proteins, inhibiting GPVI-fibrin(ogen) interaction and the tyrosine kinase inhibitors. The effect of GPVI signaling inhibitors on the procoagulant platelet number was exacerbated by eptifibatide. The tyrosine kinase inhibitors led to an increase in clot porosity; however, no differences were observed in the final clot weight, following clot retraction with the tyrosine kinase inhibitors, except for ibrutinib. In the presence of eptifibatide, clot retraction was impaired. CONCLUSION Our findings showed that GPVI-fibrin interaction significantly contributes to the development of procoagulant platelets and that inhibition of GPVI signaling increases clot porosity. Clot contractibility was impaired by the integrin αIIbβ3 and Btk pathway inhibition. Thus, inhibition of GPVI-fibrin interactions can alleviate structural characteristics that contribute to a prothrombotic clot phenotype, having potential important implications for novel antithrombotic interventions.
Collapse
Affiliation(s)
- Julia S Gauer
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Cédric Duval
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Rui-Gang Xu
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Fraser L Macrae
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Helen R McPherson
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Christian Tiede
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Darren Tomlinson
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
28
|
Zhou S, Zhao W, Hu J, Mao C, Zhou M. Application of Nanotechnology in Thrombus Therapy. Adv Healthc Mater 2023; 12:e2202578. [PMID: 36507827 DOI: 10.1002/adhm.202202578] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/26/2022] [Indexed: 12/14/2022]
Abstract
A thrombus is a blood clot that forms in the lumen of an artery or vein, restricting blood flow and causing clinical symptoms. Thrombosis is associated with many life-threatening cardiovascular diseases. However, current clinical therapeutic technologies still have many problems in targeting, enrichment, penetration, and safety to meet the thrombosis treatment needs. Therefore, researchers devote themselves to developing nanosystems loaded with antithrombotic drugs to address this paradox in recent years. Herein, the existing thrombosis treatment technologies are first reviewed; and then, their advantages and disadvantages are outlined based on a brief discussion of thrombosis's definition and formation mechanism. Furthermore, the need and application cases for introducing nanotechnology are discussed, focusing on thrombus-specific targeted ligand modification technology and microenvironment-triggered responsive drug release technology. Then, nanomaterials that can be used to design antithrombotic nanotherapeutic systems are summarized. Moreover, a variety of drug delivery technologies driven by nanomotors in thrombosis therapy is also introduced. Last of all, a prospective discussion on the future development of nanotechnology for thrombosis therapy is highlighted.
Collapse
Affiliation(s)
- Shuyin Zhou
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China.,Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Wenbo Zhao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Jinglei Hu
- Kuang Yaming Honors School, Nanjing University, Nanjing, 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Min Zhou
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| |
Collapse
|
29
|
Zhang W, Li M, Wang X, Zhang W, Wang H, Li P, Tang B. Precision Navigation of Venous Thrombosis Guided by Viscosity-Activatable Near-Infrared Fluorescence. Anal Chem 2023; 95:2382-2389. [PMID: 36653196 DOI: 10.1021/acs.analchem.2c04395] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Thrombus are blood clots formed by abnormal hemostasis in blood vessels and are closely associated with various diseases such as pulmonary embolism, myocardial infarction and stroke. Early diagnosis and treatment of thrombus is the key to reducing the high risk of thrombotic disease. Given that early thrombus is small in early size, free instability, wide regional distribution and fast formation, it is urgent to develop all-inclusive detection methods that combine high signal-to-noise ratio, in situ dynamic and rapid in-depth tissue imaging. Near-infrared (NIR) fluorescence imaging, with its excellent high spatiotemporal resolution and tissue penetration depth, is a powerful technique for direct visualization of thrombotic events in situ. Considering the fibrin highly expressed in the thrombus is a typical thrombotic target. Moreover, the viscosity of the thrombus is markedly higher than its surroundings. Therefore, we developed a fibrin-targeting and viscosity-activating thrombus NIR fluorescent probe (TIR-V) for high-resolution and high-sensitivity in situ lighten-up thrombus. TIR-V has the advantages of good thrombus targeting, significant "off-on" fluorescence specific response to viscosity, bright NIR fluorescence and good biocompatibility. The thrombus is clearly delineated by a high signal-to-noise ratio NIR fluorescence imaging, enabling imaging detection and precise navigation of thrombotic regions. This work demonstrates the potential of TIR-V as a bifunctional probe for definitive diagnostic imaging and direct navigation of thrombotic lesions in clinical applications.
Collapse
Affiliation(s)
- Wen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| | - Mengmei Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| | - Xin Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| | - Wei Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| | - Hui Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
30
|
Ali AAG, Niinuma SA, Moin ASM, Atkin SL, Butler AE. The Role of Platelets in Hypoglycemia-Induced Cardiovascular Disease: A Review of the Literature. Biomolecules 2023; 13:241. [PMID: 36830610 PMCID: PMC9953659 DOI: 10.3390/biom13020241] [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: 01/04/2023] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally as well as the leading cause of mortality and morbidity in type 2 diabetes (T2D) patients. Results from large interventional studies have suggested hyperglycemia and poor glycemic control to be largely responsible for the development of CVDs. However, the association between hypoglycemia and cardiovascular events is also a key pathophysiological factor in the development of CVDs. Hypoglycemia is especially prevalent in T2D patients treated with oral sulfonylurea agents or exogenous insulin, increasing the susceptibility of this population to cardiovascular events. The adverse cardiovascular risk of hypoglycemia can persist even after the blood glucose levels have been normalized. Hypoglycemia may lead to vascular disease through mechanisms such as enhanced coagulation, oxidative stress, vascular inflammation, endothelial dysfunction, and platelet activation. In the following review, we summarize the evidence for the role of hypoglycemia in platelet activation and the subsequent effects this may have on the development of CVD. In addition, we review current evidence for the effectiveness of therapies in reducing the risk of CVDs.
Collapse
Affiliation(s)
- Ahmed Ali Gebril Ali
- School of Medicine, Royal College of Surgeons in Ireland and Medical University of Bahrain, Busaiteen 15503, Bahrain
| | - Sara Anjum Niinuma
- School of Medicine, Royal College of Surgeons in Ireland and Medical University of Bahrain, Busaiteen 15503, Bahrain
| | - Abu Saleh Md Moin
- Research Department, Royal College of Surgeons in Ireland and Medical University of Bahrain, Busaiteen 15503, Bahrain
| | - Stephen L. Atkin
- Research Department, Royal College of Surgeons in Ireland and Medical University of Bahrain, Busaiteen 15503, Bahrain
| | - Alexandra E. Butler
- Research Department, Royal College of Surgeons in Ireland and Medical University of Bahrain, Busaiteen 15503, Bahrain
| |
Collapse
|
31
|
Ali FEM, Abd El-Aziz MK, Ali MM, Ghogar OM, Bakr AG. COVID-19 and hepatic injury: cellular and molecular mechanisms in diverse liver cells. World J Gastroenterol 2023; 29:425-449. [PMID: 36688024 PMCID: PMC9850933 DOI: 10.3748/wjg.v29.i3.425] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/15/2022] [Accepted: 12/23/2022] [Indexed: 01/12/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) represents a global health and economic challenge. Hepatic injuries have been approved to be associated with severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection. The viral tropism pattern of SARS-CoV-2 can induce hepatic injuries either by itself or by worsening the conditions of patients with hepatic diseases. Besides, other factors have been reported to play a crucial role in the pathological forms of hepatic injuries induced by SARS-CoV-2, including cytokine storm, hypoxia, endothelial cells, and even some treatments for COVID-19. On the other hand, several groups of people could be at risk of hepatic COVID-19 complications, such as pregnant women and neonates. The present review outlines and discusses the interplay between SARS-CoV-2 infection and hepatic injury, hepatic illness comorbidity, and risk factors. Besides, it is focused on the vaccination process and the role of developed vaccines in preventing hepatic injuries due to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | | | - Mahmoud M Ali
- Department of Pharmacology, Al-Azhar University, Assiut 71524, Egypt
| | - Osama M Ghogar
- Department of Biochemistry Faculty of Pharmacy, Badr University in Assiut, Egypt
| | - Adel G Bakr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| |
Collapse
|
32
|
Gaggini M, Gorini F, Vassalle C. Lipids in Atherosclerosis: Pathophysiology and the Role of Calculated Lipid Indices in Assessing Cardiovascular Risk in Patients with Hyperlipidemia. Int J Mol Sci 2022; 24:ijms24010075. [PMID: 36613514 PMCID: PMC9820080 DOI: 10.3390/ijms24010075] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The role of lipids is essential in any phase of the atherosclerotic process, which is considered a chronic lipid-related and inflammatory condition. The traditional lipid profile (including the evaluation of total cholesterol, triglycerides, high-density lipoprotein, and low-density lipoprotein) is a well-established tool to assess the risk of atherosclerosis and as such has been widely used as a pillar of cardiovascular disease prevention and as a target of pharmacological treatments in clinical practice over the last decades. However, other non-traditional lipids have emerged as possible alternative predictors of cardiometabolic risk in addition to traditional single or panel lipids, as they better reflect the overall interaction between lipid/lipoprotein fractions. Therefore, this review deals with the lipid involvement characterizing the pathophysiology of atherosclerosis, discussing some recently proposed non-traditional lipid indices and, in the light of available knowledge, their actual potential as new additive tools to better stratify cardiovascular risk in patients with hyperlipidemia as well as possible therapeutic targets in the clinical practice.
Collapse
Affiliation(s)
- Melania Gaggini
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy
| | - Francesca Gorini
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy
| | - Cristina Vassalle
- Fondazione CNR—Regione Toscana G Monasterio, Via Moruzzi 1, 56124 Pisa, Italy
- Correspondence:
| |
Collapse
|
33
|
Ye F, Zhang B, Qiu L, Zhang Y, Zhang Y, Zhang J, Zhao Q, Lu L, Zhang Z. In vivo real-time red blood cell migration and microcirculation flow synergy imaging-surveyed thrombolytic therapy with iron-oxide complexes. Mater Today Bio 2022; 16:100408. [PMID: 36097598 PMCID: PMC9463387 DOI: 10.1016/j.mtbio.2022.100408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
Nanotherapeutics as a nascent method has attracted widely interest on the treatment of thrombosis. However, due to the limited temporal and spatial resolution of conventional imaging modalities, the dynamic visualization the thrombogenesis and evaluation of the effect of thrombolytic drugs are facing severely difficulties in vivo. In addition, the development of high targeting, short circulation time, and small size thrombolysis nanotherapeutics agents requires further research. Herein, we report a synergy imaging modality that combining a label-free capillary microscopy and laser speckle microcirculation imaging, which realized dynamic visualization of single red blood cell migration and large-field dynamic blood flow. In this work, we investigated the red blood cells migration and blood flow velocity response before and after treated through introducing a functional nano-thrombolytics, iron-oxide complexes coated urokinase (IPN@UK) on an orthotopic animal model in vivo. The functionalized IPN@UK nanocomposites exhibited outstanding thrombolysis effect. Significantly, whole-course changes, including red blood cell activity, complex thrombolytic therapeutics, were well surveilled and evaluated using dual-modality combining imaging strategy. These results show this synergy imaging strategy not only can achieve multiscale non-invasive visualization of dynamic thrombus events in real-time, but also can quantify hemodynamics information of thrombus. Our study demonstrates the potential of this synergy imaging method, which for early detection of thrombus, evaluation of the effect of drug thrombolysis, developing the thrombolytic drugs, and imaging-guide thrombolytic therapy in living systems.
Collapse
Affiliation(s)
- Fei Ye
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, PR China
| | - Bei Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, 361102, PR China
| | - Lige Qiu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, PR China
| | - Yunrui Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, 361102, PR China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, 361102, PR China
| | - Jian Zhang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Department of Biomedical Engineering, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Qingliang Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, 361102, PR China
| | - Ligong Lu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, PR China
| | - Zhenlin Zhang
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, PR China
| |
Collapse
|
34
|
Zhang T, Wu S, Qin H, Wu H, Liu X, Li B, Zheng X. An Optically Controlled Virtual Microsensor for Biomarker Detection In Vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2205760. [PMID: 36074977 DOI: 10.1002/adma.202205760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/02/2022] [Indexed: 06/15/2023]
Abstract
Current technologies for the real-time analysis of biomarkers in vivo, such as needle-type microelectrodes and molecular imaging methods based on exogenous contrast agents, are still facing great challenges in either invasive detection or lack of active control of the imaging probes. In this study, by combining the design concepts of needle-type microelectrodes and the fluorescence imaging method, a new technique is developed for detecting biomarkers in vivo, named as "optically controlled virtual microsensor" (OCViM). OCViM is established by the organic integration of a specially shaped laser beam and fluorescent nanoprobe, which serve as the virtual handle and sensor tip, respectively. The laser beam can trap and manipulate the nanoprobe in a programmable manner, and meanwhile excite it to generate fluorescence emission for biosensing. On this basis, fully active control of the nanoprobe is achieved noninvasively in vivo, and multipoint detection can be realized at sub-micrometer resolution by shifting a nanoprobe among multiple positions. By using OCViM, the overexpression and heterogenous distribution of biomarkers in the thrombus is studied in living zebrafish, which is further utilized for the evaluation of antithrombotic drugs. OCViM may provide a powerful tool for the mechanism study of thrombus progression and the evaluation of antithrombotic drugs.
Collapse
Affiliation(s)
- Tiange Zhang
- Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| | - Shuai Wu
- Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| | - Haifeng Qin
- Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| | - Huaying Wu
- Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| | - Xiaoshuai Liu
- Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| | - Baojun Li
- Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| | - Xianchuang Zheng
- Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| |
Collapse
|
35
|
Brandhofer M, Hoffmann A, Blanchet X, Siminkovitch E, Rohlfing AK, El Bounkari O, Nestele JA, Bild A, Kontos C, Hille K, Rohde V, Fröhlich A, Golemi J, Gokce O, Krammer C, Scheiermann P, Tsilimparis N, Sachs N, Kempf WE, Maegdefessel L, Otabil MK, Megens RTA, Ippel H, Koenen RR, Luo J, Engelmann B, Mayo KH, Gawaz M, Kapurniotu A, Weber C, von Hundelshausen P, Bernhagen J. Heterocomplexes between the atypical chemokine MIF and the CXC-motif chemokine CXCL4L1 regulate inflammation and thrombus formation. Cell Mol Life Sci 2022; 79:512. [PMID: 36094626 PMCID: PMC9468113 DOI: 10.1007/s00018-022-04539-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 07/31/2022] [Accepted: 08/22/2022] [Indexed: 12/03/2022]
Abstract
To fulfil its orchestration of immune cell trafficking, a network of chemokines and receptors developed that capitalizes on specificity, redundancy, and functional selectivity. The discovery of heteromeric interactions in the chemokine interactome has expanded the complexity within this network. Moreover, some inflammatory mediators, not structurally linked to classical chemokines, bind to chemokine receptors and behave as atypical chemokines (ACKs). We identified macrophage migration inhibitory factor (MIF) as an ACK that binds to chemokine receptors CXCR2 and CXCR4 to promote atherogenic leukocyte recruitment. Here, we hypothesized that chemokine–chemokine interactions extend to ACKs and that MIF forms heterocomplexes with classical chemokines. We tested this hypothesis by using an unbiased chemokine protein array. Platelet chemokine CXCL4L1 (but not its variant CXCL4 or the CXCR2/CXCR4 ligands CXCL8 or CXCL12) was identified as a candidate interactor. MIF/CXCL4L1 complexation was verified by co-immunoprecipitation, surface plasmon-resonance analysis, and microscale thermophoresis, also establishing high-affinity binding. We next determined whether heterocomplex formation modulates inflammatory/atherogenic activities of MIF. Complex formation was observed to inhibit MIF-elicited T-cell chemotaxis as assessed by transwell migration assay and in a 3D-matrix-based live cell-imaging set-up. Heterocomplexation also blocked MIF-triggered migration of microglia in cortical cultures in situ, as well as MIF-mediated monocyte adhesion on aortic endothelial cell monolayers under flow stress conditions. Of note, CXCL4L1 blocked binding of Alexa-MIF to a soluble surrogate of CXCR4 and co-incubation with CXCL4L1 attenuated MIF responses in HEK293-CXCR4 transfectants, indicating that complex formation interferes with MIF/CXCR4 pathways. Because MIF and CXCL4L1 are platelet-derived products, we finally tested their role in platelet activation. Multi-photon microscopy, FLIM-FRET, and proximity-ligation assay visualized heterocomplexes in platelet aggregates and in clinical human thrombus sections obtained from peripheral artery disease (PAD) in patients undergoing thrombectomy. Moreover, heterocomplexes inhibited MIF-stimulated thrombus formation under flow and skewed the lamellipodia phenotype of adhering platelets. Our study establishes a novel molecular interaction that adds to the complexity of the chemokine interactome and chemokine/receptor-network. MIF/CXCL4L1, or more generally, ACK/CXC-motif chemokine heterocomplexes may be target structures that can be exploited to modulate inflammation and thrombosis.
Collapse
Affiliation(s)
- Markus Brandhofer
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Adrian Hoffmann
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.,Department of Anesthesiology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Xavier Blanchet
- Institute for Cardiovascular Prevention (IPEK), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Pettenkofer Straße 8a/9, 80336, Munich, Germany
| | - Elena Siminkovitch
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Anne-Katrin Rohlfing
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Omar El Bounkari
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Jeremy A Nestele
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Alexander Bild
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Christos Kontos
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), 85354, Freising, Germany
| | - Kathleen Hille
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), 85354, Freising, Germany
| | - Vanessa Rohde
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Adrian Fröhlich
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Jona Golemi
- Systems Neuroscience Group, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Ozgun Gokce
- Systems Neuroscience Group, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
| | - Christine Krammer
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Patrick Scheiermann
- Department of Anesthesiology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Nikolaos Tsilimparis
- Department of Vascular Surgery, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Nadja Sachs
- Department for Vascular and Endovascular Surgery, Klinikum Rechts Der Isar, Technische Universität München (TUM), 81675, Munich, Germany.,Munich Heart Alliance, 80802, Munich, Germany
| | - Wolfgang E Kempf
- Department for Vascular and Endovascular Surgery, Klinikum Rechts Der Isar, Technische Universität München (TUM), 81675, Munich, Germany
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum Rechts Der Isar, Technische Universität München (TUM), 81675, Munich, Germany.,Munich Heart Alliance, 80802, Munich, Germany
| | - Michael K Otabil
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Remco T A Megens
- Institute for Cardiovascular Prevention (IPEK), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Pettenkofer Straße 8a/9, 80336, Munich, Germany.,Munich Heart Alliance, 80802, Munich, Germany.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Hans Ippel
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Rory R Koenen
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Junfu Luo
- Vascular Biology and Pathology, Institute of Laboratory Medicine, Ludwig-Maximilians-Universität, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Bernd Engelmann
- Vascular Biology and Pathology, Institute of Laboratory Medicine, Ludwig-Maximilians-Universität, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Kevin H Mayo
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER, Maastricht, The Netherlands.,Department of Biochemistry, Molecular Biology and Biophysics, Health Sciences Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Aphrodite Kapurniotu
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), 85354, Freising, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Pettenkofer Straße 8a/9, 80336, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany.,Munich Heart Alliance, 80802, Munich, Germany.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Philipp von Hundelshausen
- Institute for Cardiovascular Prevention (IPEK), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Pettenkofer Straße 8a/9, 80336, Munich, Germany. .,Munich Heart Alliance, 80802, Munich, Germany.
| | - Jürgen Bernhagen
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany. .,Munich Heart Alliance, 80802, Munich, Germany.
| |
Collapse
|
36
|
Chemical composition and potential bioactivities of essential oil from Quercus mongolica bark. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
37
|
Sun S, Qiao B, Han Y, Wang B, Wei S, Chen Y. Posttranslational modifications of platelet adhesion receptors. Pharmacol Res 2022; 183:106413. [PMID: 36007773 DOI: 10.1016/j.phrs.2022.106413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/11/2022] [Accepted: 08/19/2022] [Indexed: 10/15/2022]
Abstract
Platelets play a key role in normal hemostasis, whereas pathological platelet adhesion is involved in various cardiovascular events. The underlying cause in cardiovascular events involves plaque rupture leading to subsequent platelet adhesion, activation, release, and eventual thrombosis. Traditional antithrombotic drugs often target the signal transduction process of platelet adhesion receptors by influencing the synthesis of some key molecules, and their effects are limited. Posttranslational modifications (PTMs) of platelet adhesion receptors increase the functional diversity of the receptors and affect platelet physiological and pathological processes. Antithrombotic drugs targeting PTMs of platelet adhesion receptors may represent a new therapeutic idea. In this review, various PTMs, including phosphorylation, glycosylation, ubiquitination, nitrosylation, methylation, lipidation, and proteolysis, of three platelet adhesion receptors, glycoprotein Ib-IX-V (GPIb-IX-V), glycoprotein VI (GPVI), and integrin αIIbβ3, are reviewed. It is important to comprehensively understand the PTMs process of platelet adhesion receptors.
Collapse
Affiliation(s)
- Shukun Sun
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Bao Qiao
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yu Han
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Bailu Wang
- Clinical Trial Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Shujian Wei
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China.
| | - Yuguo Chen
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
38
|
Li Q, Xie E, Tu Y, Wu Y, Guo Z, Li P, Li Y, Yu X, Ye Z, Yu C, Gao Y, Jingang Z. Association between kaolin-induced maximum amplitude and slow-flow/no-reflow in ST elevation myocardial infarction patients treated with primary percutaneous coronary intervention. Int J Cardiol 2022; 369:13-18. [PMID: 35970443 DOI: 10.1016/j.ijcard.2022.08.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/08/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND ST-segment elevation myocardial infarction (STEMI) patients with a high thrombus burden have a relatively high slow-flow/no-reflow risk. However, the association between kaolin-induced maximum amplitude (MAthrombin) and slow-flow/no-reflow has been scarcely explored. METHODS STEMI patients treated with primary percutaneous coronary intervention (PCI) were retrospectively enrolled from January 2015 to December 2019 at China-Japan Friendship Hospital. MAthrombin levels were measured using thromboelastography before the PCI procedure. The patients were divided into two groups according to thrombolysis in myocardial infarction (TIMI) flow grade after primary PCI: the normal flow group (TIMI flow grade = 3) and slow-flow/no-reflow (TIMI flow grade ≤ 2). The logistic regression model and restricted cubic spline regression (RCS) were used to analyze the predictive value of MAthrombin for slow-flow/no-reflow. All patients were followed up after discharge and observed the adverse cardiovascular events between the two groups. RESULTS A total of 690 patients were enrolled, with 108(15.7%) having slow-flow/no-reflow. The multivariate logistic regression model analysis showed that MAthrombin level was an independent risk factor for slow-flow/no-reflow. The RCS analysis showed a nonlinear relationship between MAthrombin levels and slow-flow/no-reflow. The cut-off value of MAthrombin levels for predicting slow-flow/no-reflow was 68 mm. During a median follow-up time of 4.4 years, slow-flow/no-reflow (hazard ratio 1.93, 95% confidence interval 1.27-2.93, P = 0.002) and MAthrombin levels (hazard ratio 1.06, 95% confidence interval 1.03-1.08, P < 0.001) were independent risk factors for predicting the long-term of adverse clinical cardiovascular events. CONCLUSION MAthrombin was an independent risk factor for predicting slow-flow/ no-reflow in STEMI patients who underwent primary PCI.
Collapse
Affiliation(s)
- Qing Li
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China; Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Enmin Xie
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yimin Tu
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yaxin Wu
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China; Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Ziyu Guo
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China; Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Peizhao Li
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China; Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yike Li
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xiaozhai Yu
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Zixiang Ye
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China; Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Changan Yu
- Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yanxiang Gao
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China; Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Zheng Jingang
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China.
| |
Collapse
|
39
|
Tang X, Manamanchaiyaporn L, Zhou Q, Huang C, Li L, Li Z, Wang L, Wang J, Ren L, Xu T, Yan X, Zheng Y. Synergistic Integration and Pharmacomechanical Function of Enzyme-Magnetite Nanoparticle Swarms for Low-Dose Fast Thrombolysis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2202848. [PMID: 35905497 DOI: 10.1002/smll.202202848] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/08/2022] [Indexed: 06/15/2023]
Abstract
Magnetic micro-/nanoparticles are extensively explored over the past decade as active diagnostic/therapeutic agents for minimally invasive medicine. However, sufficient function integration on these miniaturized bodies toward practical applications remains challenging. This work proposes a synergistic strategy via integrating particle functionalization and bioinspired swarming, demonstrated by recombinant tissue plasminogen activator modified magnetite nanoparticles (rtPA-Fe3 O4 NPs) for fast thrombolysis in vivo with low drug dosage. The synthesized rtPA-Fe3 O4 NPs exhibit superior magnetic performance, high biocompatibility, and thrombolytic enzyme activity. Benefiting from a customized magnetic operation system designed for animal experiments and preclinical development, these agglomeration-free NPs can assemble into micro-/milli-scale swarms capable of robust maneuver and reconfigurable transformation for on-demand tasks in complex biofluids. Specifically, the spinning mode of the swarm exerts focused fluid shear stresses while rubbing on the thrombus surface, constituting a mechanical force for clot breakdown. The synergy of the NPs' inherent enzymatic effect and swarming-triggered fluid forces enables amplified efficacy of thrombolysis in an in vivo occlusion model of rabbit carotid artery, using lower drug concentration than clinical dosage. Furthermore, swarming-enhanced ultrasound signals aid in imaging-guided treatment. Therefore, the pharmacomechanical NP swarms herein represent an injectable thrombolytic tool joining advantages of intravenous drug therapy and robotic intervention.
Collapse
Affiliation(s)
- Xiuzhen Tang
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, 200233, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361005, China
- Center of Excellence in Creative Engineering Design and Development & Department of Mechanical Engineering, Faculty of Engineering, Thammasat University, Pathumthani, 12121, Thailand
| | - Laliphat Manamanchaiyaporn
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361005, China
- Center of Excellence in Creative Engineering Design and Development & Department of Mechanical Engineering, Faculty of Engineering, Thammasat University, Pathumthani, 12121, Thailand
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Qi Zhou
- School of Engineering, Institute for Multiscale Thermofluids, University of Edinburgh, Edinburgh, EH9 3FD, UK
| | - Chenyang Huang
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lihuang Li
- Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China
| | - Ziqiao Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361005, China
| | - Longchen Wang
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, 200233, China
| | - Jienan Wang
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, 200233, China
| | - Lei Ren
- Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China
| | - Tiantian Xu
- Center of Excellence in Creative Engineering Design and Development & Department of Mechanical Engineering, Faculty of Engineering, Thammasat University, Pathumthani, 12121, Thailand
| | - Xiaohui Yan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361005, China
| | - Yuanyi Zheng
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, 200233, China
| |
Collapse
|
40
|
Qiao Y, Zhang K, Zhang Z, Zhang C, Sun Y, Feng Z. Fermented soybean foods: A review of their functional components, mechanism of action and factors influencing their health benefits. Food Res Int 2022; 158:111575. [PMID: 35840260 DOI: 10.1016/j.foodres.2022.111575] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/27/2022]
Abstract
After thousands of years of evolution and development, traditional fermented soybean foods, with their unique charm, have gained a stable place in the global market. With the explosive development of modern biological technologies, some traditional fermented soybean foods that possess health-promoting benefits are gradually appearing. Physiologically active substances in fermented soybean foods have received extensive attention in recent decades. This review addresses the potential health benefits of several representative fermented soybean foods, as well as the action mechanism and influencing factors of their functional components. Phenolic compounds, low-molecular-weight peptides, melanoidins, furanones and 3-hydroxyanthranilic acid are the antioxidative components predominantly found in fermented soybean foods. Angiotensin I-converting enzyme inhibitory peptides and γ-aminobutyric acid isolated from fermented soy foods provide potential selectivity for hypertension therapy. The potential anti-inflammatory bioactive components in fermented soybean foods include γ-linolenic acid, butyric acid, soy sauce polysaccharides, 2S albumin and isoflavone glycones. Deoxynojirimycin, genistein, and betaine possess high activity against α-glucosidase. Additionally, fermented soybean foods contain neuroprotective constituents, including indole alkaloids, nattokinase, arbutin, and isoflavone vitamin B12. The anticancer activities of fermented soybean foods are associated with surfactin, isolavone, furanones, trypsin inhibitors, and 3-hydroxyanthranilic acid. Nattokinase is highly correlated with antioxidant activity. And a high level of menaquinones-7 is linked to protection against neurodegenerative diseases. Sufficiently recognizing and exploiting the health benefits and functional components of traditional fermented soybean foods could provide a new strategy in the development of the food fermentation industry.
Collapse
Affiliation(s)
- Yali Qiao
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No.600, Changjiang Road, Harbin 150030, China
| | - Kenan Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No.600, Changjiang Road, Harbin 150030, China
| | - Zongcai Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No.600, Changjiang Road, Harbin 150030, China
| | - Chao Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No.600, Changjiang Road, Harbin 150030, China
| | - Yan Sun
- Heilongjiang Tobacco Industry Co., Ltd. Harbin Cigarette Factory, Harbin 150027, China
| | - Zhen Feng
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No.600, Changjiang Road, Harbin 150030, China; Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wanning 571533, China.
| |
Collapse
|
41
|
Pancaldi F, Kim OV, Weisel JW, Alber M, Xu Z. Computational Biomechanical Modeling of Fibrin Networks and Platelet-Fiber Network Interactions. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022; 22:100369. [PMID: 35386550 PMCID: PMC8979495 DOI: 10.1016/j.cobme.2022.100369] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fibrin deformation and interaction of fibrin with other blood components play critical roles in hemostasis and thrombosis. In this review, computational and mathematical biomechanical models of fibrin network deformation and contraction at different spatio-temporal scales as well as challenges in developing and calibrating multiscale models are discussed. There are long standing challenges. For instance, applicability of models to identify and test potential mechanisms of the biomechanical processes mediating interactions between platelets and fiber networks in blood clot stretching and contraction needs to be examined carefully. How the structural and mechanical properties of major blood clot components influences biomechanical responses of the entire clot subjected to external forces, such as blood flow or vessel wall deformations needs to be investigated thoroughly.
Collapse
Affiliation(s)
- Francesco Pancaldi
- Department of Mathematics, University of California Riverside, Riverside, CA 92505, USA
- Center for Quantitative Modeling in Biology, University of California Riverside, Riverside, CA 92505, USA
| | - Oleg V. Kim
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - John W. Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Mark Alber
- Department of Mathematics, University of California Riverside, Riverside, CA 92505, USA
- Center for Quantitative Modeling in Biology, University of California Riverside, Riverside, CA 92505, USA
| | - Zhiliang Xu
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, IN 46556, USA
| |
Collapse
|
42
|
Keller K, Prochaska JH, Coldewey M, Göbel S, Schmitt VH, Hahad O, Ullmann A, Nagler M, Lamparter H, Espinola-Klein C, Münzel T, Wild PS. Atherosclerosis and Its Impact on the Outcomes of Patients with Deep Venous Thrombosis. Life (Basel) 2022; 12:734. [PMID: 35629401 PMCID: PMC9143312 DOI: 10.3390/life12050734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 01/10/2023] Open
Abstract
Introduction: Atherosclerosis and pulmonary embolism (PE) affect cardiovascular mortality substantially. We aimed to investigate the impact of atherosclerosis on the outcomes of patients with deep venous thrombosis (DVT) and to identify the differences in DVT patients with and without PE. Methods: Patients with DVT with and without symptomatic atherosclerosis (defined as coronary artery disease, myocardial infarction and/or peripheral artery disease) as well as with and without PE under oral anticoagulation were enrolled during January 2011−April 2013 and compared. The impact of symptomatic atherosclerosis on several outcomes was analyzed. Results: Overall, 509 DVT patients (70.0 [56.0−77.0] years, 51.9% females) were included in this study. Among them, 179 (36.3%) had symptomatic atherosclerosis and 204 (40.1%) a concomitant PE. DVT patients with symptomatic atherosclerosis were older (74.0 [IQR 65.0−80.0] vs. 63.0 [48.0−75.0] years, p < 0.0001), more often male (56.4% vs. 43.9%, p = 0.0087) and had a higher prevalence of classical CVRF and a higher Charlson comorbidity index (7.00 [5.00−8.00] vs. 4.00 [2.00−6.00], p < 0.001). Symptomatic atherosclerosis was associated with increased mortality (HR 1.98 [95%CI 1.12−3.49], p = 0.018) and hospitalizations (HR 1.64 [95%CI 1.21−2.21], p = 0.0012) and primary long-term outcome (HR 1.99 [95%CI 1.31−3.04], p = 0.0013) during the 2 years follow-up-period in DVT patients. DVT patients without PE had diabetes mellitus (28.2% vs. 16.3%, p < 0.01) and symptomatic atherosclerosis (42.9% vs. 26.4%, p < 0.001) more often compared to DVT patients with PE, and symptomatic atherosclerosis was associated with isolated DVT (without PE) (OR 2.01 [95%CI 1.28−3.16], p < 0.01). Conclusions: Atherosclerosis was associated with isolated DVT (without PE) and increased mortality in DVT patients under oral anticoagulation. The profile of CVRF and comorbidities differed between DVT patients with and without a concomitant PE. In the case of DVT or PE, patients should be screened for concomitant atherosclerotic disease. Clinical Trial Registration: at clinicaltrials with Unique identifier NCT01809015.
Collapse
Affiliation(s)
- Karsten Keller
- Department of Cardiology, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (M.C.); (S.G.); (V.H.S.); (O.H.); (C.E.-K.); (T.M.)
- Center for Thrombosis and Hemostasis, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (J.H.P.); (A.U.); (M.N.); (H.L.); (P.S.W.)
- Department of Sports Medicine, Medical Clinic VII, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Jürgen H. Prochaska
- Center for Thrombosis and Hemostasis, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (J.H.P.); (A.U.); (M.N.); (H.L.); (P.S.W.)
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, 55131 Mainz, Germany
| | - Meike Coldewey
- Department of Cardiology, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (M.C.); (S.G.); (V.H.S.); (O.H.); (C.E.-K.); (T.M.)
- Center for Thrombosis and Hemostasis, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (J.H.P.); (A.U.); (M.N.); (H.L.); (P.S.W.)
| | - Sebastian Göbel
- Department of Cardiology, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (M.C.); (S.G.); (V.H.S.); (O.H.); (C.E.-K.); (T.M.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, 55131 Mainz, Germany
| | - Volker H. Schmitt
- Department of Cardiology, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (M.C.); (S.G.); (V.H.S.); (O.H.); (C.E.-K.); (T.M.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, 55131 Mainz, Germany
| | - Omar Hahad
- Department of Cardiology, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (M.C.); (S.G.); (V.H.S.); (O.H.); (C.E.-K.); (T.M.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, 55131 Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), 55131 Mainz, Germany
| | - Alexander Ullmann
- Center for Thrombosis and Hemostasis, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (J.H.P.); (A.U.); (M.N.); (H.L.); (P.S.W.)
| | - Markus Nagler
- Center for Thrombosis and Hemostasis, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (J.H.P.); (A.U.); (M.N.); (H.L.); (P.S.W.)
| | - Heidrun Lamparter
- Center for Thrombosis and Hemostasis, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (J.H.P.); (A.U.); (M.N.); (H.L.); (P.S.W.)
| | - Christine Espinola-Klein
- Department of Cardiology, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (M.C.); (S.G.); (V.H.S.); (O.H.); (C.E.-K.); (T.M.)
| | - Thomas Münzel
- Department of Cardiology, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (M.C.); (S.G.); (V.H.S.); (O.H.); (C.E.-K.); (T.M.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, 55131 Mainz, Germany
| | - Philipp S. Wild
- Center for Thrombosis and Hemostasis, University Medical Center Mainz of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (J.H.P.); (A.U.); (M.N.); (H.L.); (P.S.W.)
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, 55131 Mainz, Germany
| |
Collapse
|
43
|
Engineered extracellular vesicles and their mimics in cardiovascular diseases. J Control Release 2022; 347:27-43. [PMID: 35508222 DOI: 10.1016/j.jconrel.2022.04.046] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 01/08/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. Current pharmacological interventions for the CVDs suffer from low bioavailability, low retention rate, poor targeting, drug resistance complicated side effects. Extracellular vesicles (EVs), which are lipid vesicles secreted by cells, play key roles in pathological processes of CVDs. Engineered EVs and EV mimics with superior properties can overcome limitations of traditional medicine, thus emerging as alternative therapeutic options for the CVDs. In this Review, we summarized basic concepts of EVs and EV mimics, highlighted engineering strategies, and lastly discussed applications of engineered EVs and EV mimics against the CVDs. We believe this Review can provide some new insights on engineering EVs and EV mimics and facilitate their application in precise control of CVDs.
Collapse
|
44
|
Angiolillo DJ, Galli M, Collet JP, Kastrati A, O'Donoghue ML. Antiplatelet therapy after percutaneous coronary intervention. EUROINTERVENTION 2022; 17:e1371-e1396. [PMID: 35354550 PMCID: PMC9896394 DOI: 10.4244/eij-d-21-00904] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/16/2021] [Indexed: 11/23/2022]
Abstract
Antiplatelet therapy is key to reducing local thrombotic complications and systemic ischaemic events among patients undergoing percutaneous coronary interventions (PCI), but it is inevitably associated with increased bleeding. The continuous refinement in stent technologies, together with the high incidence of ischaemic recurrences after PCI and the understanding of prognostic implications associated with bleeding, have led to a substantial evolution in antiplatelet treatment regimens over the past decades. Numerous investigations have been conducted to better stratify patients undergoing PCI according to their ischaemic and bleeding risks and to implement antithrombotic regimens accordingly. Evidence from these investigations have resulted in a number of antithrombotic treatment options as recommended by recent guidelines. In this State-of-the-Art review we provide the rationale, summarise the evidence, and discuss current and future directions of antiplatelet treatment regimens after PCI.
Collapse
Affiliation(s)
- Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Mattia Galli
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Jean-Philippe Collet
- ACTION Study Group, Institut de Cardiologie, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Adnan Kastrati
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Michelle L O'Donoghue
- TIMI Study Group, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
45
|
Nanocarrier-Based Management of Venous and Arterial Thrombosis. CRYSTALS 2022. [DOI: 10.3390/cryst12040450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Cardiovascular diseases represent the leading cause of mortality worldwide, with recent epidemiological studies revealing an increasing trend of prevalence and incidence globally. Among cardiovascular disorders, both arterial and venous thrombosis and particularly their acute life-threating complications such as ischemic stroke, acute myocardial infarction, deep venous thrombosis and pulmonary embolism are responsible for more than 25% of all deaths worldwide. The modern approach following progresses in anticoagulant, thrombolytic and antiaggregant therapies has significantly improved the prognoses of these conditions in the last past decades. However, several challenges still remain such as achieving the optimal drug concentration at the injured site, reducing the shortcomings of drug resistance and the incidence of life-threatening hemorrhages. Nanomedicine is a well-known field of medicine in which atomic and molecular structures ranging between 0.1–100 nm are used in various domains due to their specific mechanical, electrical, thermal and magnetic properties. Recent experimental and clinical evidence have shown that nanotechnology could be a safe, effective and an appealing approach for various non-cardiovascular and cardiovascular diseases such as thromboembolic conditions. In this review, we have described the most promising nanotechnology-based approaches not only for the diagnosis, but also for the treatment of vascular thrombotic diseases.
Collapse
|
46
|
D’Ardes D, Boccatonda A, Cocco G, Fabiani S, Rossi I, Bucci M, Guagnano MT, Schiavone C, Cipollone F. Impaired coagulation, liver dysfunction and COVID-19: Discovering an intriguing relationship. World J Gastroenterol 2022; 28:1102-1112. [PMID: 35431501 PMCID: PMC8985482 DOI: 10.3748/wjg.v28.i11.1102] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 08/09/2021] [Accepted: 02/15/2022] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is, at present, one of the most relevant global health problems. In the literature hepatic alterations have been described in COVID-19 patients, and they are mainly represented by worsening of underlying chronic liver disease leading to hepatic decompensation and liver failure with higher mortality. Several potential mechanisms used by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to cause liver damage have been hypothesized. COVID-19 primary liver injury is less common than secondary liver injury. Most of the available data demonstrate how liver damage in SARS-CoV-2 infection is likely due to systemic inflammation, and it is less likely mediated by a cytopathic effect directed on liver cells. Moreover, liver alterations could be caused by hypoxic injury and drugs (antibiotics and non-steroidal anti-inflammatory drugs, remdesivir, tocilizumab, tofacitinib and dexamethasone). SARS-CoV-2 infection can induce multiple vascular district atherothrombosis by affecting simultaneously cerebral, coronary and peripheral vascular beds. Data in the literature highlight how the virus triggers an exaggerated immune response, which added to the cytopathic effect of the virus can induce endothelial damage and a prothrombotic dysregulation of hemostasis. This leads to a higher incidence of symptomatic and confirmed venous thrombosis and of pulmonary embolisms, especially in central, lobar or segmental pulmonary arteries, in COVID-19. There are currently fewer data for arterial thrombosis, while myocardial injury was identified in 7%-17% of patients hospitalized with SARS-CoV-2 infection and 22%-31% in the intensive care unit setting. Available data also revealed a higher occurrence of stroke and more serious forms of peripheral arterial disease in COVID-19 patients. Hemostasis dysregulation is observed during the COVID-19 course. Lower platelet count, mildly increased prothrombin time and increased D-dimer are typical laboratory features of patients with severe SARS-CoV-2 infection, described as "COVID-19 associated coagulopathy." These alterations are correlated to poor outcomes. Moreover, patients with severe SARS-CoV-2 infection are characterized by high levels of von Willebrand factor with subsequent ADAMTS13 deficiency and impaired fibrinolysis. Platelet hyperreactivity, hypercoagulability and hypofibrinolysis during SARS-CoV-2 infection induce a pathological state named as "immuno-thromboinflammation." Finally, liver dysfunction and coagulopathy are often observed at the same time in patients with COVID-19. The hypothesis that liver dysfunction could be mediated by microvascular thrombosis has been supported by post-mortem findings and extensive vascular portal and sinusoidal thrombosis observation. Other evidence has shown a correlation between coagulation and liver damage in COVID-19, underlined by the transaminase association with coagulopathy, identified through laboratory markers such as prothrombin time, international normalized ratio, fibrinogen, D-dimer, fibrin/fibrinogen degradation products and platelet count. Other possible mechanisms like immunogenesis of COVID-19 damage or massive pericyte activation with consequent vessel wall fibrosis have been suggested.
Collapse
Affiliation(s)
- Damiano D’Ardes
- “Clinica Medica” Institute, Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Andrea Boccatonda
- Unit of Ultrasound, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Giulio Cocco
- Unit of Ultrasound, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Stefano Fabiani
- Unit of Ultrasound, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Ilaria Rossi
- “Clinica Medica” Institute, Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Marco Bucci
- “Clinica Medica” Institute, Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Maria Teresa Guagnano
- “Clinica Medica” Institute, Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Cosima Schiavone
- Unit of Ultrasound, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Francesco Cipollone
- “Clinica Medica” Institute, Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| |
Collapse
|
47
|
Role of NETosis in Central Nervous System Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3235524. [PMID: 35028005 PMCID: PMC8752220 DOI: 10.1155/2022/3235524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/18/2021] [Indexed: 11/17/2022]
Abstract
Central nervous system (CNS) injury is divided into brain injury and spinal cord injury and remains the most common cause of morbidity and mortality worldwide. Previous reviews have defined numerous inflammatory cells involved in this process. In the human body, neutrophils comprise the largest numbers of myeloid leukocytes. Activated neutrophils release extracellular web-like DNA amended with antimicrobial proteins called neutrophil extracellular traps (NETs). The formation of NETs was demonstrated as a new method of cell death called NETosis. As the first line of defence against injury, neutrophils mediate a variety of adverse reactions in the early stage, and we consider that NETs may be the prominent mediators of CNS injury. Therefore, exploring the specific role of NETs in CNS injury may help us shed some light on early changes in the disease. Simultaneously, we discovered that there is a link between NETosis and other cell death pathways by browsing other research, which is helpful for us to establish crossroads between known cell death pathways. Currently, there is a large amount of research concerning NETosis in various diseases, but the role of NETosis in CNS injury remains unknown. Therefore, this review will introduce the role of NETosis in CNS injury, including traumatic brain injury, cerebral ischaemia, CNS infection, Alzheimer's disease, and spinal cord injury, by describing the mechanism of NETosis, the evidence of NETosis in CNS injury, and the link between NETosis and other cell death pathways. Furthermore, we also discuss some agents that inhibit NETosis as therapies to alleviate the severity of CNS injury. NETosis may be a potential target for the treatment of CNS injury, so exploring NETosis provides a feasible therapeutic option for CNS injury in the future.
Collapse
|
48
|
Mukherjee AK, Chattopadhyay DJ. Potential clinical applications of phytopharmaceuticals for the in-patient management of coagulopathies in COVID-19. Phytother Res 2022; 36:1884-1913. [PMID: 35147268 PMCID: PMC9111032 DOI: 10.1002/ptr.7408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 12/21/2022]
Abstract
Thrombotic complications occur in many cardiovascular pathologies and have been demonstrated in COVID‐19. The currently used antithrombotic drugs are not free of adverse reactions, and COVID‐19 patients in particular, when treated with a therapeutic dose of an anticoagulant do not receive mortality benefits. The clinical management of COVID‐19 is one of the most difficult tasks for clinicians, and the search for safe, potent, and effective antithrombotic drugs may benefit from exploring naturally bioactive molecules from plant sources. This review describes recent advances in understanding the antithrombotic potential of herbal drug prototypes and points to their future clinical use as potent antithrombotic drugs. Although natural products are perceived to be safe, their clinical and therapeutic applications are not always apparent or accepted. More in‐depth studies are necessary to demonstrate the clinical usefulness of plant‐derived, bioactive compounds. In addition, holistic approaches in systematic investigations and the identification of antithrombotic mechanisms of the herbal bioactive molecule(s) need to be conducted in pre‐clinical studies. Moreover, rigorous studies are needed to compare the potency of herbal drugs to that of competitor chemical antithrombotic drugs, and to examine their interactions with Western antithrombotic medicines. We have also proposed a road map to improve the commercialization of phytopharmaceuticals.
Collapse
Affiliation(s)
- Ashis K Mukherjee
- Division of Life Sciences, Institute of Advanced Study in Science and Technology, Guwahati, India.,Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, India
| | | |
Collapse
|
49
|
Li J, Shang C, Rong Y, Sun J, Cheng Y, He B, Wang Z, Li M, Ma J, Fu B, Ji X. Review on Laser Technology in Intravascular Imaging and Treatment. Aging Dis 2022; 13:246-266. [PMID: 35111372 PMCID: PMC8782552 DOI: 10.14336/ad.2021.0711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/11/2021] [Indexed: 12/14/2022] Open
Abstract
Blood vessels are one of the most essential organs, which nourish all tissues in our body. Once there are intravascular plaques or vascular occlusion, other organs and circulatory systems will not work properly. Therefore, it is necessary to detect abnormal blood vessels by intravascular imaging technologies for subsequent vascular treatment. The emergence of lasers and fiber optics promotes the development of intravascular imaging and treatment. Laser imaging techniques can obtain deep vascular images owing to light scattering and absorption properties. Moreover, photothermal and photomechanical effects of laser make it possible to treat vascular diseases accurately. In this review, we present the research progress and applications of laser techniques in intravascular imaging and treatment. Firstly, we introduce intravascular optical coherent tomography and intravascular photoacoustic imaging, which can obtain various information of plaques. Multimodal intravascular imaging techniques provide more information about intravascular plaques, which have an essential influence on intravascular imaging. Secondly, two laser techniques including laser angioplasty and endovenous laser ablation are discussed for the treatment of arterial and venous diseases, respectively. Finally, the outlook of laser techniques in blood vessels, as well as the integration of laser imaging and treatment are prospected in the section of discussions.
Collapse
Affiliation(s)
- Jing Li
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Ce Shang
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Yao Rong
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
- Medical Engineering Devices of Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Jingxuan Sun
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
| | - Yuan Cheng
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
| | - Boqu He
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
| | - Zihao Wang
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
| | - Ming Li
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Jianguo Ma
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
| | - Bo Fu
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China.
- Key Laboratory of Big Data-Based Precision Medicine Ministry of Industry and Information Technology, Interdisciplinary Innovation Institute of Medicine and Engineering, Beihang University, Beijing, China.
| | - Xunming Ji
- BUAA-CCMU Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Neurosurgery Department of Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
50
|
THE EFFECTS OF THYROID-STIMULATING HORMONE, FREE THYROXINE LEVELS, AND THYROID ANTIBODIES ON MEAN PLATELET VOLUME: ORIGINAL RESEARCH. JOURNAL OF CONTEMPORARY MEDICINE 2022. [DOI: 10.16899/jcm.932053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|