1
|
Mora Massad K, Dai Z, Petrache I, Ventetuolo CE, Lahm T. Lung endothelial cell heterogeneity in health and pulmonary vascular disease. Am J Physiol Lung Cell Mol Physiol 2025; 328:L877-L884. [PMID: 39772753 DOI: 10.1152/ajplung.00296.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/16/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
Lung endothelial cells (ECs) are essential for maintaining organ function and homeostasis. Despite sharing some common features with ECs from organ systems, lung ECs exhibit significant heterogeneity in morphology, function, and gene expression. This heterogeneity is increasingly recognized as a key contributor to the development of pulmonary diseases like pulmonary hypertension (PH). In this mini-review, we explore the evolving understanding of lung EC heterogeneity, particularly through the lens of single-cell RNA sequencing (scRNA-seq) technologies. These advances have provided unprecedented insights into the diverse EC subpopulations, their specific roles, and the disturbances in their homeostatic functions that contribute to PH pathogenesis. In particular, these studies identified novel and functionally distinct cell types such as aerocytes and general capillary ECs that are critical for maintaining lung function in health and disease. In addition, multiple novel pathways and mechanisms have been identified that contribute to aberrant pulmonary vascular remodeling in PH. Emerging techniques like single-nucleus RNA sequencing and spatial transcriptomics have further pushed the field forward by discovering novel disease mediators. As research continues to leverage these advanced techniques, the field is poised to uncover novel EC subtypes and disease mechanisms, paving the way for new therapeutic targets in PH and other lung diseases.
Collapse
Grants
- 4IPA1275127 American Heart Association (AHA)
- Reuben M. Chernaick Fellowship
- R01HL169509 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Borstein Family Foundation
- R01HL144727 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- I01 BX002042 BLRD VA
- Colorado Pulmonary Vascular Disease Award
- R01 HL170096 NHLBI NIH HHS
- R01HL170096 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL158596 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL077328 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01 HL158507 NHLBI NIH HHS
- R01 HL169509 NHLBI NIH HHS
- R01 HL158596 NHLBI NIH HHS
- R01 HL162794 NHLBI NIH HHS
- R01-HL141268 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL62794 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- Karina Mora Massad
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Zhiyu Dai
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine in St. Louis, Saint Louis, Missouri, United States
| | - Irina Petrache
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, United States
| | - Corey E Ventetuolo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States
- Department of Health Services, Policy and Practice, Brown University, Providence, Rhode Island, United States
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, United States
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado, United States
| |
Collapse
|
2
|
Du Z, Hu X, Lin Y, Chen L, Huang Y, Fan J, Yang S. Applications of microfluidic chip technology in microvascular thrombosis research. Mikrochim Acta 2025; 192:371. [PMID: 40413353 DOI: 10.1007/s00604-025-07239-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 05/11/2025] [Indexed: 05/27/2025]
Abstract
The formation and progression of microvascular thrombosis are critical mechanisms underlying many vascular-related diseases. Therefore, replicating the microvascular blood flow environment in vitro and investigating the mechanisms of microvascular thrombosis formation are highly significant. In recent years, microfluidic chip technology has been extensively applied in in vitro research for its capability to systematically and comprehensively replicate the complex processes of microvascular thrombosis in laboratory settings. This review systematically examines the development and applications of microfluidic chip technology in microvascular thrombosis research. It begins with a brief summary of the technical features of microfluidic chip technology, followed by a detailed discussion of its applications in constructing in vitro microvascular models, investigating thrombosis mechanisms, and evaluating antithrombotic drug efficacy. Finally, the review summarizes the current research progress and discusses potential directions for future development. This review also systematically explains the breakthrough contribution of microfluidic chips from the perspective of engineering bionics and provides new insights for the pathological research and clinical management of microvascular thrombosis: constructing a high-precision physiological simulation system through bionic topology design and dynamic fluid regulation to achieve high-precision reconstruction of vascular dynamic microenvironment; based on the systems-level dynamics analysis, the dynamic evolution law of multi-factor synergy in the process of thrombosis is revealed; construct a drug-response evaluation system and establish a transformation bridge from micro-mechanism to clinical intervention. In summary, this review is expected to accelerate the development of targeted therapies and diagnostic tools for microvascular thrombosis.
Collapse
Affiliation(s)
- Zhichang Du
- College of Marine Equipment and Mechanical Engineering, Jimei University, Xiamen, 361021, China.
| | - Xiaolong Hu
- College of Marine Equipment and Mechanical Engineering, Jimei University, Xiamen, 361021, China
| | - Yurui Lin
- College of Marine Equipment and Mechanical Engineering, Jimei University, Xiamen, 361021, China
| | - Ling Chen
- College of Marine Equipment and Mechanical Engineering, Jimei University, Xiamen, 361021, China
| | - Yan Huang
- College of Marine Equipment and Mechanical Engineering, Jimei University, Xiamen, 361021, China
| | - Jianyu Fan
- College of Marine Equipment and Mechanical Engineering, Jimei University, Xiamen, 361021, China
| | - Shaohui Yang
- College of Marine Equipment and Mechanical Engineering, Jimei University, Xiamen, 361021, China.
- Key Laboratory of Ocean Renewable Energy Equipment of Fujian Province, Xiamen, 361021, China.
- Key Laboratory of Energy Cleaning Utilizationand, Development of Fujian Province , Xiamen, 361021, China.
| |
Collapse
|
3
|
Lv CL, Li B. Interface morphodynamics in living tissues. SOFT MATTER 2025; 21:3670-3687. [PMID: 40226989 DOI: 10.1039/d5sm00145e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Interfaces between distinct tissues or between tissues and environments are common in multicellular organisms. The evolution and stability of these interfaces are essential for tissue development, and their dysfunction can lead to diseases such as cancer. Mounting efforts, either theoretical or experimental, have been devoted to uncovering the morphodynamics of tissue interfaces. Here, we review the recent progress of studies on interface morphodynamics. The regulatory mechanisms governing interface evolution are dissected, with a focus on adhesion, cortical tension, cell activity, extracellular matrix, and microenvironment. We examine the methodologies used to study morphodynamics, emphasizing the characteristics of experimental techniques and theoretical models. Finally, we explore the broader implications of interface morphodynamics in tissue morphogenesis and diseases, offering a comprehensive perspective on this rapidly developing field.
Collapse
Affiliation(s)
- Cheng-Lin Lv
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing, China.
| | - Bo Li
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing, China.
- Mechano-X Institute, Department of Engineering Mechanics, Tsinghua University, Beijing, China
- State Key Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, China
| |
Collapse
|
4
|
Ma YN, Ma SR, Yang L, Wu J, Wang YR, Bao LJ, Ma L, Wu QQ, Wang ZH. Diagnostic biomarkers and immune infiltration profiles common to COVID-19, acute myocardial infarction and acute ischaemic stroke using bioinformatics methods and machine learning. BMC Neurol 2025; 25:201. [PMID: 40340571 PMCID: PMC12060493 DOI: 10.1186/s12883-025-04212-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/28/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND COVID-19 is a disease that affects people globally. Beyond affecting the respiratory system, COVID-19 patients are at an elevated risk for both venous and arterial thrombosis. This heightened risk contributes to an increased probability of acute complications, including acute myocardial infarction (AMI) and acute ischemic stroke (AIS). Given the unclear relationship between COVID-19, AMI, and AIS, it is crucial to gain a deeper understanding of their associations and potential molecular mechanisms. This study aims to utilize bioinformatics to analyze gene expression data, identify potential therapeutic targets and biomarkers, and explore the role of immune cells in the disease. METHODS This study employed three Gene Expression Omnibus (GEO) datasets for analysis, which included data on COVID-19, AMI and AIS. We performed enrichment analysis on the co-DEGs for these three diseases to clarify gene pathways and functions, and also examined the relationship between co-DEGs and immune infiltration. Machine learning techniques and protein-protein interaction networks (PPI) were used to identify hub genes within the co-DEGs. Finally, we employed a dual validation strategy integrating independent GEO datasets and in vitro experiments with human blood samples to comprehensively assess the reliability of our experimental findings. RESULTS We identified 88 co-DEGs associated with COVID-19, AMI and AIS. Enrichment analysis results indicated that co-DEGs were significantly enriched in immune inflammatory responses related to leukocytes and neutrophils. Immune infiltration analysis revealed significant differences in immune cell populations between the disease group and the normal group. Finally, genes selected through machine learning methods included: CLEC4E, S100A12, and IL1R2. Based on the PPI network, the top ten most influential DEGs were identified as MMP9, TLR2, TLR4, ITGAM, S100A12, FCGR1A, CD163, FCER1G, FPR2, and CLEC4D. The integration of the protein-protein interaction (PPI) network with machine learning techniques facilitated the identification of S100A12 as a potential common biomarker for early diagnosis and a therapeutic target for all three diseases. Ultimately, validation of S100A12 showed that it was consistent with our experimental results, confirming its reliability as a biomarker. Moreover, it demonstrated good diagnostic performance for the three diseases. CONCLUSION We employed bioinformatics methods and machine learning to investigate common diagnostic biomarkers and immune infiltration characteristics of COVID-19, AMI and AIS. Functional and pathway analyses indicated that the co-DEGs were primarily enriched in immune inflammatory responses related to leukocytes and neutrophils. Through two machine learning approaches and the PPI network, and subsequent validation and evaluation, we identified S100A12 as a potential common therapeutic target and biomarker related to immune response that may influence these three diseases.
Collapse
Affiliation(s)
- Ya-Nan Ma
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Si-Rong Ma
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Li Yang
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Juan Wu
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ya-Rong Wang
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Li-Jia Bao
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Li Ma
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Qing-Qiu Wu
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Zhen-Hai Wang
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.
- Diagnosis and Treatment Engineering Technology Research Center of Nervous System Diseases of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China.
- Neurology Center, Ningxia Medical University General Hospital, Yinchuan, Ningxia, China.
| |
Collapse
|
5
|
Vanderkamp SG, Niazy M, Stegelmeier AA, Stinson KJ, Ricker N, Bridle BW. Cytokine, chemokine, and acute-phase protein profiles in plasma as correlative biomarkers of clinical outcomes for patients with COVID-19. Sci Rep 2025; 15:15397. [PMID: 40316702 PMCID: PMC12048561 DOI: 10.1038/s41598-025-99248-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 04/17/2025] [Indexed: 05/04/2025] Open
Abstract
Coronavirus disease identified in 2019 (COVID-19), caused by severe acute respiratory syndrome-coronavirus-2, had a global impact on human health and the economy. The aim of this study was to quantify cytokines, chemokines, and acute phase proteins in the plasma of patients with COVID-19 to elucidate potential biomarkers to inform prognostic and treatment decisions. Clustering analysis using the K-prototypes method identified underlying biological patterns in patients with COVID-19. The penalized multinomial logistic regression analysis identified two comorbidities (hypertension, congestive heart failure) and thirteen analytes as potential risk factors for COVID-19 progression with 88.2% accuracy. Based on a patient's age, high concentrations of interleukin (IL)-6, monocyte chemoattractant protein-1, and pentraxin 3 were important biomarkers for lethal COVID-19. Decreased concentrations of interferon gamma-induced protein-10, IL-10, and soluble tumor necrosis factor receptor I were found to be associated with mild COVID-19, while increasing concentrations of these analytes could be used to predict COVID-19 severity.
Collapse
Affiliation(s)
- Sierra G Vanderkamp
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Maysa Niazy
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Ashley A Stegelmeier
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | | | - Nicole Ricker
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
6
|
Joy S, Prasannan A, Venkatachalam K, Binesh A. Molecular Mechanism of Notch Signaling and Macrophages in Deep Vein Thrombosis: A Comprehensive Review. Cell Biochem Biophys 2025:10.1007/s12013-025-01761-y. [PMID: 40279070 DOI: 10.1007/s12013-025-01761-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
Deep vein thrombosis is an acute medical condition, and the molecular basis of this etiology will be crucial in the discovery of more advanced therapies. This review has focused at the Notch signaling pathway, which plays a significant role in different physiological activities such as homeostasis, development, and disease. Also, reveal macrophage function in inflammation and thrombosis in depth, with a focus on their polarization and interaction with the endothelium during thrombosis. In this context, some essential cellular and molecular mechanisms relevant to thrombus pathogenesis, DVT aetiology and risk factors, as well as elements and composition of the Notch pathway, are covered in the end, with a focus on elements that distinguish canonical from non-canonical signaling pathways and their biological relevance to macrophages. Notch signaling has been shown to influence macrophage activation and polarization, influencing their function in thrombosis breakdown and resolution. This interplay between Notch signaling and macrophages may reveal possible treatment targets for DVT. Discuss the physiological role of Notch signaling in vascular biology, as well as how it contributes to thrombosis. The difficulties in implementing these discoveries in clinical practice are discussed, along with the status of ongoing clinical trials and experimental investigations focussing on macrophage-directed treatments and Notch inhibitors. These molecular insights synthesis provides a basis for the creation of novel strategies for the efficient management of DVT.
Collapse
Affiliation(s)
- Sisira Joy
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, Tamil Nadu, India
| | - Anusha Prasannan
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, Tamil Nadu, India
| | - Kaliyamurthi Venkatachalam
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, Tamil Nadu, India
| | - Ambika Binesh
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, Tamil Nadu, India.
| |
Collapse
|
7
|
Ni J, Cao X, Hu X, Fu S, Jiang M, Ni Y. Okra polysaccharide mitigates carrageenan-induced thrombosis in mice by regulating inflammation and oxidative stress. Front Pharmacol 2025; 16:1576108. [PMID: 40276603 PMCID: PMC12018850 DOI: 10.3389/fphar.2025.1576108] [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: 02/14/2025] [Accepted: 03/19/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction Thrombosis is a serious health hazard, which has been paid more and more attention.Okra polysaccharide (OP) is a biologically active substance extracted from okra which exhibits anti-inflammation and anti-oxidative properties. Nevertheless, the effect of OP on thrombosis is still unknown. In this study, we determined whether OP can suppress carrageenan-induced mice thrombosis and its involved mechanism. Methods Twenty-four BALB/c mice were assigned to four groups randomly (6 mice/group): Ctrl, Model, OP low lose (OP-L, 200 mg/kg body weight), and OP high lose (OP-H,400 mg/kg body weight) were administered via intragastric administration for 9 days. Tails were photographed before collecting for H&E and Masson staining. Liver and lung tissues were collected for H&E staining, RT-qPCR, Western blot and GSH content detection. Injury or dysfunction of endothelial cells (ECs) was assessed using RT-qPCR, Western blot and cell adhesion assays. Results OP can effectively improve carrageenan-induced thrombosis in tissues of mice (tail, liver, and lung) in vivo. In addition, OP inhibited inflammation by suppressing the toll-like receptor 4 (TLR4)/nuclear factor κB (NF-κB) pathway and reduced oxidative damage by elevating the level of GSH and antioxidant enzyme in liver and lung tissues. In vitro, OP inhibited thrombin-induced human platelet clots retraction, and decreased lipopolysaccharide (LPS)-activated adhesion of THP-1 monocytes to human umbilical vein endothelial cells(HUVECs) by suppressing intercellular adhesion molecule-1 (ICAM-1) level. Discussion In conclusion, OP can inhibit thrombosis in mouse model by regulating inflammation and oxidative stress, which suggest that OP could act as a potential functional food for prevention of thrombus.
Collapse
Affiliation(s)
- Jinping Ni
- Department of Cardiology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xiwen Cao
- The QUEEN MARY School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xianqing Hu
- Department of Cardiology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
- Department of Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Shenwen Fu
- Department of Cardiology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
- The QUEEN MARY School, Jiangxi Medical College, Nanchang University, Nanchang, China
- Department of Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Meixiu Jiang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuqi Ni
- Department of Cardiology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
- Department of Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
8
|
Hergueta-Redondo M, Sánchez-Redondo S, Hurtado B, Santos V, Pérez-Martínez M, Ximénez-Embún P, McDowell SAC, Mazariegos MS, Mata G, Torres-Ruiz R, Rodríguez-Perales S, Martínez L, Graña-Castro O, Megias D, Quail D, Quintela-Fandino M, Peinado H. The impact of a high fat diet and platelet activation on pre-metastatic niche formation. Nat Commun 2025; 16:2897. [PMID: 40175356 PMCID: PMC11965330 DOI: 10.1038/s41467-025-57938-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/07/2025] [Indexed: 04/04/2025] Open
Abstract
There is active crosstalk between tumor cells and the tumor microenvironment during metastatic progression, a process that is significantly affected by obesity, particularly in breast cancer. Here we analyze the impact of a high fat diet (HFD) on metastasis, focusing on the role of platelets in the formation of premetastatic niches (PMNs). We find that a HFD provokes pre-activation of platelets and endothelial cells, promoting the formation of PMNs in the lung. These niches are characterized by increased vascular leakiness, platelet activation and overexpression of fibronectin in both platelets and endothelial cells. A HFD promotes interactions between platelets, tumor cells and endothelial cells within PMNs, enhancing tumor cell homing and metastasis. Importantly, therapeutic interventions like anti-platelet antibody administration or a dietary switch reduce metastatic cell homing and outgrowth. Moreover, blocking fibronectin reduces the interaction of tumor cells with endothelial cells. Importantly, when coagulation parameters prior to neoadjuvant treatment are considered, triple negative breast cancer (TNBC) female patients with reduced Partial Thromboplastin time (aPTT) had a significantly shorter time to relapse. These findings highlight how diet and platelet activation in pre-metastatic niches affect tumor cell homing and metastasis, suggesting potential therapeutic interventions and prognostic markers for TNBC patients.
Collapse
Affiliation(s)
- Marta Hergueta-Redondo
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Sara Sánchez-Redondo
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Begoña Hurtado
- Cancer Cell Cycle Group, Preclinical & Translational Research Department, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Vanesa Santos
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Manuel Pérez-Martínez
- Confocal Microscopy Unit, Biotechnology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Pilar Ximénez-Embún
- Proteomics Unit, Biotechnology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Sheri A C McDowell
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Marina S Mazariegos
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund Stem Cell Center (SCC), Lund University, Lund, Sweden
| | - Gadea Mata
- Confocal Microscopy Unit, Biotechnology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Department of Mathematics and Computer Science, University of La Rioja, La Rioja, Spain
| | - Raúl Torres-Ruiz
- Molecular Cytogenetics Unit, Human Cancer Genetics Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Division of Hematopoietic Innovative Therapies, Biomedical Innovation Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnologicas (CIEMAT), Madrid, Spain
- Advanced Therapies Unit, Instituto de Investigacion Sanitaria Fundacion Jiménez Díaz, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Sandra Rodríguez-Perales
- Molecular Cytogenetics Unit, Human Cancer Genetics Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Lola Martínez
- Flow Cytometry Core Unit, Biotechnology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Osvaldo Graña-Castro
- Bioinformatics Unit, Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA-Nemesio Díez), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, 28925, Alcorcón, Spain
| | - Diego Megias
- Confocal Microscopy Unit, Biotechnology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Advanced Optical Microscopy - ISCIII Madrid, Madrid, Spain
| | - Daniela Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Miguel Quintela-Fandino
- Breast Cancer Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
- Medical Oncology, Hospital de Fuenlabrada, Madrid, Spain
| | - Héctor Peinado
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain.
| |
Collapse
|
9
|
Masset C, Drillaud N, Ternisien C, Degauque N, Gerard N, Bruneau S, Branchereau J, Blancho G, Mesnard B, Brouard S, Giral M, Cantarovich D, Dantal J. The concept of immunothrombosis in pancreas transplantation. Am J Transplant 2025; 25:650-668. [PMID: 39709128 DOI: 10.1016/j.ajt.2024.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/06/2024] [Accepted: 11/23/2024] [Indexed: 12/23/2024]
Abstract
Early failure of a pancreatic allograft due to complete thrombosis has an incidence of approximately 10% and is the main cause of comorbidity in pancreas transplantation. Although several risk factors have been identified, the exact mechanisms leading to this serious complication are still unclear. In this review, we define the roles of the individual components involved during sterile immunothrombosis-namely endothelial cells, platelets, and innate immune cells. Further, we review the published evidence linking the main risk factors for pancreatic thrombosis to cellular activation and vascular modifications. We also explore the unique features of the pancreas itself: the vessel endothelium, specific vascularization, and relationship to other organs-notably the spleen and adipose tissue. Finally, we summarize the therapeutic possibilities for the prevention of pancreatic thrombosis depending on the different mechanisms such as anticoagulation, anti-inflammatory molecules, endothelium protectors, antagonism of damage-associated molecular patterns, and use of machine perfusion.
Collapse
Affiliation(s)
- Christophe Masset
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France.
| | - Nicolas Drillaud
- Laboratory of Hemostasis, Nantes University Hospital, Nantes, France
| | | | - Nicolas Degauque
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Nathalie Gerard
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Sarah Bruneau
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Julien Branchereau
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Gilles Blancho
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Benoit Mesnard
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Sophie Brouard
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Magali Giral
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Diego Cantarovich
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Jacques Dantal
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| |
Collapse
|
10
|
Cacciatore S, Andaloro S, Bernardi M, Oterino Manzanas A, Spadafora L, Figliozzi S, Asher E, Rana JS, Ecarnot F, Gragnano F, Calabrò P, Gallo A, Andò G, Manzo-Silberman S, Roeters van Lennep J, Tosato M, Landi F, Biondi-Zoccai G, Marzetti E, Sabouret P. Chronic Inflammatory Diseases and Cardiovascular Risk: Current Insights and Future Strategies for Optimal Management. Int J Mol Sci 2025; 26:3071. [PMID: 40243756 PMCID: PMC11989023 DOI: 10.3390/ijms26073071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Chronic inflammation is a pivotal driver in the progression of atherosclerosis, significantly contributing to the burden of cardiovascular disease (CVD). Patients with chronic inflammatory diseases, such as inflammatory bowel diseases (IBDs) (e.g., ulcerative colitis and Crohn's disease), rheumatological disorders, as well as individuals with auto-immune diseases (such as systemic lupus erythematosus), present a higher risk of major adverse cardiac events (MACEs). Despite their elevated CVD risk, these populations remain underrepresented in cardiovascular research, leading to a critical underestimation of their cardiovascular risk (CVR) in clinical practice. Furthermore, even recent CVR scores poorly predict the risk of events in these specific populations. This narrative review examines the physiopathological mechanisms linking chronic inflammation, immunomodulation, atherosclerosis, thrombosis and cardiovascular events. We review data from epidemiological studies and clinical trials to explore the potential cardiovascular benefits of anti-inflammatory and immunomodulatory therapies. Despite existing evidence, significant gaps in knowledge remain. Future research is mandatory, focusing on innovative strategies for risk stratification and optimization, including lipidomics, proteomics, advanced inflammatory markers, microbiota profiling, and cardiovascular imaging. Addressing these unmet needs will enhance understanding of cardiovascular risk in chronic inflammatory diseases, enabling tailored interventions and better outcomes.
Collapse
Affiliation(s)
- Stefano Cacciatore
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy;
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
| | - Silvia Andaloro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy;
| | - Marco Bernardi
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy; (M.B.); (L.S.); (G.B.-Z.)
| | - Armando Oterino Manzanas
- Department of Cardiology, Hospital Universitario de Salamanca-IBSAL, Paseo de San Vicente, 58-182, 37007 Salamanca, Spain;
| | - Luigi Spadafora
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy; (M.B.); (L.S.); (G.B.-Z.)
| | - Stefano Figliozzi
- IRCCS Humanitas Research Hospital, Via Alessandro Manzoni, 56, Rozzano, 20089 Milano, Italy;
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, Pieve Emanuele, 20090 Milano, Italy
| | - Elad Asher
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Shmuel (Hans) Beyth St. 12, Jerusalem 9103102, Israel;
| | - Jamal S. Rana
- Division of Cardiology, Kaiser Permanente Northern California, 1 Kaiser Plaza, Oakland, CA 94612, USA;
- Division of Research, Kaiser Permanente Northern California, 1 Kaiser Plaza, Oakland, CA 94612, USA
| | - Fiona Ecarnot
- Department of Cardiology, University Hospital, Boulevard Fleming, 25000 Besançon, France;
- SINERGIES Unit, University Marie & Louis Pasteur, 19 Rue Ambroise Paré, 25000 Besançon, France
| | - Felice Gragnano
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Via Leonardo Bianchi, Ospedale Monaldi, 80131 Naples, Italy; (F.G.); (P.C.)
- Division of Cardiology, A.O.R.N. “Sant’Anna e San Sebastiano”, Via Ferdinando Palasciano, 81100 Caserta, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Via Leonardo Bianchi, Ospedale Monaldi, 80131 Naples, Italy; (F.G.); (P.C.)
- Division of Cardiology, A.O.R.N. “Sant’Anna e San Sebastiano”, Via Ferdinando Palasciano, 81100 Caserta, Italy
| | - Antonio Gallo
- INSERM UMR1166, IHU ICAN, Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, Pitié-Salpêtrière Hospital, Sorbonne University, AP-HP, 47–83 Bd de l’Hôpital, 75013 Paris, France;
| | - Giuseppe Andò
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “Gaetano Martino”, Via Consolare Valeria, 1, 98124 Messina, Italy;
| | - Stephane Manzo-Silberman
- ACTION Study Group, Inserm UMRS1166, Heart Institute, Pitié-Salpetriere Hospital, Sorbonne University, 47-83 Bd de l’Hôpital, 75013 Paris, France; (S.M.-S.); (P.S.)
| | - Jeanine Roeters van Lennep
- Department of Internal Medicine, Cardiovascular Institute, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands;
| | - Matteo Tosato
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
| | - Francesco Landi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy;
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy; (M.B.); (L.S.); (G.B.-Z.)
- Maria Cecilia Hospital, GVM Care & Research, Via Corriera, 1, 48033 Cotignola, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy;
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
| | - Pierre Sabouret
- ACTION Study Group, Inserm UMRS1166, Heart Institute, Pitié-Salpetriere Hospital, Sorbonne University, 47-83 Bd de l’Hôpital, 75013 Paris, France; (S.M.-S.); (P.S.)
| |
Collapse
|
11
|
He J, Chen Y, Li Y, Feng Y. Molecular mechanisms and therapeutic interventions in acute kidney injury: a literature review. BMC Nephrol 2025; 26:144. [PMID: 40121405 PMCID: PMC11929251 DOI: 10.1186/s12882-025-04077-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
Acute kidney injury (AKI) is a clinical challenge characterized by elevated morbidity and a substantial impact on individual health and socioeconomic factors. A comprehensive examination of the molecular pathways behind AKI is essential for its prevention and management. In recent years, vigorous research in the domain of AKI has concentrated on pathophysiological characteristics, early identification, and therapeutic approaches across many aetiologies and highlighted the principal themes of oxidative stress, inflammatory response, apoptosis, necrosis, and immunological response. This review comprehensively reviewed the molecular mechanisms underlying AKI, including oxidative stress, inflammatory pathways, immune cell-mediated injury, activation of the renin-angiotensin-aldosterone (RAAS) system, mitochondrial damage and autophagy, apoptosis, necrosis, etc. Inflammatory pathways are involved in the injuries in all four structural components of the kidney. We also summarized therapeutic techniques and pharmacological agents associated with the aforementioned molecular pathways. This work aims to clarify the molecular mechanisms of AKI thoroughly, offer novel insights for further investigations of AKI, and facilitate the formulation of efficient therapeutic methods to avert the progression of AKI.
Collapse
Affiliation(s)
- Jiajia He
- Department of Nephrology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yanqin Chen
- Department of Nephrology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yi Li
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Chengdu, 610072, China
| | - Yunlin Feng
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Chengdu, 610072, China.
| |
Collapse
|
12
|
Abou Mansour M, El Rassi C, Sleem B, Borghol R, Arabi M. Thromboembolic Events in the Era of COVID-19: A Detailed Narrative Review. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2025; 2025:3804576. [PMID: 40226433 PMCID: PMC11986918 DOI: 10.1155/cjid/3804576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/14/2025] [Indexed: 04/15/2025]
Abstract
COVID-19, caused by the SARS-CoV-2 virus, is not only characterized by respiratory symptoms but is also associated with a wide range of systemic complications, including significant hematologic abnormalities. This is a comprehensive review of the current literature, using PubMed and Google Scholar, on the pathophysiology and incidence of thromboembolic events in COVID-19 patients and thromboprophylaxis. COVID-19 infection induces a prothrombotic state in patients through the dysregulation of the renin-angiotensin-aldosterone system (RAAS), endothelial dysfunction, elevated von Willebrand factor (vWF), and a dysregulated immune response involving the complement system and neutrophil extracellular traps (NETs). As a result, thromboembolic complications have emerged in COVID-19 cases, occurring more frequently in severe cases and hospitalized patients. These thrombotic events affect both venous and arterial circulation, with increased incidences of deep venous thrombosis (DVT), pulmonary embolism (PE), systemic arterial thrombosis, and myocardial infarction (MI). While DVT and PE are more common, the literature highlights the potential lethal consequences of arterial thromboembolism (ATE). This review also briefly examines the ongoing discussions regarding the use of anticoagulants for the prevention of thrombotic events in COVID-19 patients. While theoretically promising, current studies have yielded varied outcomes: Some suggest potential benefits, whereas others report an increased risk of bleeding events among hospitalized patients. Therefore, further large-scale studies are needed to assess the efficacy and safety of anticoagulants for thromboprophylaxis in COVID-19 patients.
Collapse
Affiliation(s)
- Maria Abou Mansour
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Christophe El Rassi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Bshara Sleem
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Raphah Borghol
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, Division of Pediatric Hematology-Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mariam Arabi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, Division of Pediatric Cardiology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
13
|
Dobariya KH, Goyal D, Kumar H. Molecular signature-based labeling techniques for vascular endothelial cells. Acta Histochem 2025; 127:152222. [PMID: 39644518 DOI: 10.1016/j.acthis.2024.152222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 11/14/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
Vascular endothelial cells (VECs) play a crucial role in the development and maintenance of vascular biology specific to the tissue types. Molecular signature-based labeling and imaging of VECs help researchers understand potential mechanisms linking VECs to disease pathology, serving as valuable biomarkers in clinical settings and trials. Labeling techniques involve selectively tagging or marking VECs for visualization. Immunolabeled employs antibodies that specifically bind to VECs markers, while fluorescent tracers or dyes can directly label VECs for imaging. Some techniques use specific carbohydrate residues on cell surface, while others employ endothelial-specific promoters to express fluorescent proteins. Additionally, VEC can be labeled with contrast agents, radiolabeled tracers, and nanoparticles. The choice of labeling technique depends on study context, including whether it involves animal models, in vitro cell cultures, or clinical applications. Herein, we discussed the various labeling methods utilized to label VECs and the techniques to visualize them.
Collapse
Affiliation(s)
- Krutika H Dobariya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Divya Goyal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
14
|
Chang JJ, Brew K, Hamilton JA, Kumar V, Diaz JA, Takayama S. Bioprinted Micro-Clots for Kinetic Analysis of Endothelial Cell-Mediated Fibrinolysis. Adv Healthc Mater 2025; 14:e2403043. [PMID: 39887618 PMCID: PMC11912099 DOI: 10.1002/adhm.202403043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 01/17/2025] [Indexed: 02/01/2025]
Abstract
Vascular hypo-fibrinolysis is a historically underappreciated and understudied aspect of venous thromboembolism (VTE). This paper describes the development of a micro-clot dissolution assay for quantifying the fibrinolytic capacity of endothelial cells - a key driver of VTE development. This assay is enabled using aqueous two-phase systems (ATPS) to bioprint microscale fibrin clots over human umbilical vein endothelial cells (HUVECs). Importantly, these micro-clots are orders of magnitude smaller than conventional fibrin constructs and allow HUVEC-produced plasminogen activators to mediate visually quantifiable fibrinolysis. Using live-cell time-lapse imaging, micro-clot dissolution by HUVECs is tracked, and fibrinolysis kinetics are quantified. The sensitivity of cell-driven fibrinolysis to various stimuli is rapidly tested. The physiological relevance of this convenient high-throughput assay is illustrated through treatments with lipopolysaccharide (LPS) and rosuvastatin that elicit anti- and pro-fibrinolytic responses, respectively. Furthermore, treatment with baricitinib, an anti-inflammatory therapeutic found to increase cardiovascular risks after market approval, provokes an anti-fibrinolytic response - which highlights the potential role of endothelial cells in increasing VTE risk for patients receiving this drug. This endothelial cell fibrinolysis assay provides a high-throughput and versatile drug testing platform - potentially allowing for early preclinical identification of therapeutics that may beneficially enhance or adversely impair endothelial fibrinolysis.
Collapse
Affiliation(s)
- Jonathan J. Chang
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
- The Parker H. Petit Institute of Bioengineering and BioscienceGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Kelsey Brew
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Jamie A.G. Hamilton
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Varun Kumar
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - José A. Diaz
- Division of Surgical ResearchVanderbilt University Medical CenterNashvilleTN37232USA
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
- The Parker H. Petit Institute of Bioengineering and BioscienceGeorgia Institute of TechnologyAtlantaGA30332USA
| |
Collapse
|
15
|
Piccirillo F, Lanciotti M, Nusca A, Frau L, Spanò A, Liporace P, Ussia GP, Grigioni F. Sodium-Glucose Transporter-2 Inhibitors (SGLT2i) and Myocardial Ischemia: Another Compelling Reason to Consider These Agents Regardless of Diabetes. Int J Mol Sci 2025; 26:2103. [PMID: 40076724 PMCID: PMC11899902 DOI: 10.3390/ijms26052103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
In recent years, the introduction of sodium-glucose transporter-2 inhibitors (SGLT2is) marked a significant advancement in the treatment of cardiovascular disease (CVD). Beyond their known effects on glycemic control and lipid profile, SGLT2is demonstrate notable benefits for cardiovascular morbidity and mortality, regardless of diabetic status. These agents are currently recommended as first-line therapies in patients with heart failure, both with reduced and preserved ejection fraction, as they improve symptoms and reduce the risk of hospitalization. While several studies have demonstrated that SGLT2is can reduce the incidence of major adverse cardiovascular events (MACEs), the true impact of these agents on atherosclerosis progression and myocardial ischemia remains to be fully understood. A global beneficial effect related to improved glycemic and lipid control could be hypothesized, even though substantial evidence shows a direct impact on molecular pathways that enhance endothelial function, exhibit anti-inflammatory properties, and provide myocardial protection. In this context, this narrative review summarizes the current knowledge regarding these novel anti-diabetic drugs in preventing and treating myocardial ischemia, aiming to define an additional area of application beyond glycemic control and heart failure.
Collapse
Affiliation(s)
- Francesco Piccirillo
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (M.L.); (L.F.); (A.S.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Matteo Lanciotti
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (M.L.); (L.F.); (A.S.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Annunziata Nusca
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (M.L.); (L.F.); (A.S.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Lorenzo Frau
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (M.L.); (L.F.); (A.S.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Agostino Spanò
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (M.L.); (L.F.); (A.S.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Paola Liporace
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (M.L.); (L.F.); (A.S.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Gian Paolo Ussia
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (M.L.); (L.F.); (A.S.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Francesco Grigioni
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (M.L.); (L.F.); (A.S.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| |
Collapse
|
16
|
Subramaniam S, Jose A, Kenney D, O’Connell AK, Bosmann M, Douam F, Crossland N. Challenging the notion of endothelial infection by SARS-CoV-2: insights from the current scientific evidence. Front Immunol 2025; 16:1443932. [PMID: 39967675 PMCID: PMC11832389 DOI: 10.3389/fimmu.2025.1443932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 01/14/2025] [Indexed: 02/20/2025] Open
Affiliation(s)
- Saravanan Subramaniam
- Department of Pharmacology and Toxicology, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
- Renal Section, Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Asha Jose
- Renal Section, Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Devin Kenney
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Aoife K. O’Connell
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Markus Bosmann
- Department of Medicine, Pulmonary Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Florian Douam
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Nicholas Crossland
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| |
Collapse
|
17
|
Dugbartey GJ, Penney LN, Mills L, Zhang MY, Juriasingani S, Major S, McLeod P, Liu W, Haig A, Wood ME, Torregrossa R, Whiteman M, Turley E, Postenka C, Sener A. AP39, a novel mitochondria-targeted hydrogen sulfide donor, promotes cutaneous wound healing in an in vivo murine model of acute frostbite injury. Biomed Pharmacother 2025; 183:117869. [PMID: 39879854 DOI: 10.1016/j.biopha.2025.117869] [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: 11/18/2024] [Revised: 01/16/2025] [Accepted: 01/23/2025] [Indexed: 01/31/2025] Open
Abstract
Frostbite injury refers to cold tissue injury which typically affects the peripheral areas of the body, and is associated with limb loss and high rates of morbidity. Historically, treatment options have been limited to supportive care, leading to suboptimal outcomes for affected patients. The pathophysiology of frostbite injury has been understood in recent years to share similarity with that of cold ischemia-reperfusion injury as seen in solid organ transplantation, of which mitochondria play an important contributing role. The present study investigated whether AP39, a novel mitochondria-targeted slow-releasing hydrogen sulfide donor, applied topically in a vehicle cream at 200 nM or 1 µM could mitigate frostbite injury and promote wound healing in mice. Frostbite injury was induced continuously for 3 min on the dorsal skin of C57BL/6 mice (Mus musculus) using magnets frozen on dry ice (-80 °C). AP39, delivered via a vehicle cream, was used daily to treat frostbite injury until animals were euthanized on day 15 after induction of frostbite injury. Wound tissues were stained with hematoxylin and eosin along with immunofluorescence staining with cleaved caspase-3, CD31, KI-67, CD163, fibronectin and cytokeratin. While 200 nM AP39 improved granulation tissue maturation (p < 0.001), angiogenesis (p < 0.01) and cell proliferation (p < 0.001) compared to vehicle control, 1 µM AP39 further increased granulation tissue formation compared to other frostbite groups (p < 0.001). Thus, AP39 promoted frostbite wound healing, and therefore could be considered as a treatment option for patients with frostbite injury.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Surgery, Division of Urology, London Health Sciences Center, Western University, London, Ontario, Canada; Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, Ontario, Canada; Multi-Organ Transplant Program, London Health Sciences Center, Western University, London, Ontario, Canada; Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana; Department of Physiology and Pharmacology, Accra College of Medicine, East Legon, Accra, Ghana
| | - Lucas N Penney
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, Ontario, Canada; Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Lauren Mills
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, Ontario, Canada; Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Max Y Zhang
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, Ontario, Canada; Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Smriti Juriasingani
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, Ontario, Canada; Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Sally Major
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, Ontario, Canada
| | - Patrick McLeod
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, Ontario, Canada
| | - Winnie Liu
- Department of Pathology, Schulich School of Medicine & Dentistry, University of Western, London, Ontario, Canada
| | - Aaron Haig
- Department of Pathology, Schulich School of Medicine & Dentistry, University of Western, London, Ontario, Canada
| | - Mark E Wood
- St. Luke's Campus, University of Exeter Medical School, Exeter EX1 2LU, UK
| | | | - Matthew Whiteman
- St. Luke's Campus, University of Exeter Medical School, Exeter EX1 2LU, UK
| | - Eva Turley
- Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Carl Postenka
- London Regional Cancer Program, London Health Sciences Centre, Western University, London, Ontario, Canada
| | - Alp Sener
- Department of Surgery, Division of Urology, London Health Sciences Center, Western University, London, Ontario, Canada; Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, Ontario, Canada; Multi-Organ Transplant Program, London Health Sciences Center, Western University, London, Ontario, Canada; Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
18
|
Deal H, Byrnes EM, Pandit S, Sheridan A, Brown AC, Daniele M. Injury-on-a-chip for modelling microvascular trauma-induced coagulation. LAB ON A CHIP 2025; 25:440-453. [PMID: 39763291 PMCID: PMC11704661 DOI: 10.1039/d4lc00471j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025]
Abstract
Blood coagulation is a highly regulated injury response that features polymerization of fibrin fibers to prevent the passage of blood from a damaged vascular endothelium. A growing body of research seeks to monitor coagulation in microfluidic systems but fails to capture coagulation as a response to disruption of the vascular endothelium. Here we present a device that allows compression injury of a defined segment of a microfluidic vascular endothelium and the assessment of coagulation at the injury site. This pressure injury-on-a-chip (PINCH) device allows visualization of coagulation as the accumulation of fluorescent fibrin at injury sites. Quantification of fluorescent fibrin levels upstream of and at injury sites confirm that pre-treating vascular endothelium with fluid shear stress helps capture coagulation as an injury response. We leverage the PINCH devices to demonstrate the limited coagulation response of type A hemophiliacs and evaluate the performance of hemostatic microparticles and fibrinolytic nanoparticles. Our findings and the straightforward fabrication of the PINCH devices make it a promising choice for additional screening of hemostatic therapeutics.
Collapse
Affiliation(s)
- Halston Deal
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 1840 Entrepreneur Dr., Raleigh, NC, 27695 USA.
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA.
| | - Elizabeth M Byrnes
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 1840 Entrepreneur Dr., Raleigh, NC, 27695 USA.
| | - Sanika Pandit
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 1840 Entrepreneur Dr., Raleigh, NC, 27695 USA.
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA.
| | - Anastasia Sheridan
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 1840 Entrepreneur Dr., Raleigh, NC, 27695 USA.
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA.
| | - Ashley C Brown
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 1840 Entrepreneur Dr., Raleigh, NC, 27695 USA.
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA.
| | - Michael Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 1840 Entrepreneur Dr., Raleigh, NC, 27695 USA.
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA.
- Department of Electrical & Computer Engineering, North Carolina State University, 890 Oval Dr., Raleigh, NC, 27695 USA
| |
Collapse
|
19
|
Asahara N, Ebisu H, Yuki S, Fujita R, Kojima S. Paraoxonase 1 ameliorates neurological symptoms and motor coordination impairment caused by cerebral ischemia-reperfusion injury. Biomed Pharmacother 2025; 182:117792. [PMID: 39733589 DOI: 10.1016/j.biopha.2024.117792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/13/2024] [Accepted: 12/21/2024] [Indexed: 12/31/2024] Open
Abstract
The anti-atherosclerotic effects of high-density lipoprotein (HDL) prevent the onset of cerebral infarction and provide cerebroprotective effects against ischemia-reperfusion injury. These inhibitory effects have been attributed to its antioxidant, anti-inflammatory, and antithrombotic properties. However, pharmacotherapeutic strategies to clinically realize these effects have not been demonstrated. Therefore, we aimed to develop paraoxonase 1 (PON1), a hydrolytic enzyme associated with HDL that exhibits antioxidant and anti-inflammatory effects, as a novel therapeutic agent against ischemia-reperfusion injury in cerebral infarction. We established a method to extract PON1 from human plasma with high purity and recovery while maintaining its activity. The purified PON1 exhibited antioxidant activity against human-derived LDL and HDL. Furthermore, PON1 actively suppressed the oxidation chain reaction by hydrolyzing lipid peroxides. The HDL-binding ability of PON1 was evaluated based on its activity in fractionated HDL from mice administered PON1 intravenously, which showed that most intravenously administered PON1 specifically bound to HDL. The cerebroprotective effect of intravenously administered PON1 was assessed using a mouse middle cerebral artery ischemia-reperfusion model by measuring infarct volume, long-term neurological scores, and walking time on a rotarod. Administration of PON1 after reperfusion reduced infarct volume 24 h after ischemia-reperfusion. Additionally, daily administration of PON1 for three days significantly improved neurological scores and walking time by approximately one month. Analysis of gene arrays in brain tissue indicated that PON1 suppresses biological functions and pathways associated with oxidative stress, inflammation, vascular dysfunction, thrombosis, and fibrosis. PON1 enhances the cerebroprotective effects of HDL and is a potential candidate for acute stroke therapy.
Collapse
Affiliation(s)
- Naomi Asahara
- Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama-shi, Kanagawa 227-0033, Japan.
| | - Hajime Ebisu
- Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama-shi, Kanagawa 227-0033, Japan.
| | - Satoshi Yuki
- Development & Medical Affairs Division, Mitsubishi Tanabe Pharma Corporation, 1-1-1, Marunouchi Chiyoda-ku, Tokyo 100-8205, Japan.
| | - Ryo Fujita
- Research Division, Mitsubishi Tanabe Pharma Corporation, Shonan Health Innovation Park, 2-26-1, Muraoka-Higashi, Fujisawa-shi, Kanagawa 251-8555, Japan.
| | - Shinji Kojima
- Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama-shi, Kanagawa 227-0033, Japan.
| |
Collapse
|
20
|
Zavoriti A, Miossec P. Understanding Cardiovascular Events With JAK Inhibitors: Tofacitinib Reduces Synovial and Vascular Inflammation but not the Prothrombotic Effects of Inflammatory Cytokines on Endothelium. ACR Open Rheumatol 2025; 7:e11790. [PMID: 39800890 PMCID: PMC11725533 DOI: 10.1002/acr2.11790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 10/30/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
OBJECTIVE Inflammation drives cardiovascular disease in rheumatoid arthritis (RA). Treatment with tofacitinib, a JAK1/JAK3 inhibitor, is associated with increased cardiovascular events in patients with RA. Here, we determined its effects on cytokine production during interactions between immune cells at the synovial and vascular levels and its impact on endothelial activation and coagulation during inflammation. METHODS Activated human peripheral blood mononuclear cells (PBMCs) were cocultured with RA synoviocytes or endothelial cells (ECs) mimicking the cellular interactions in synovium and vessels responsible for cytokine production. A dose-response of tofacitinib was tested on interferon γ, interleukin (IL) 17A, IL-10, IL-6, and IL-1β, and cytokine production was measured by enzyme-linked immunosorbent assay at 48 hours. Endothelial activation was induced with IL-17A and tumor necrosis factor (TNF) or on contact with PBMCs. Shortly after tofacitinib treatment, the expression of EC activation markers, specifically IL-6, IL-8, vascular cell adhesion molecule 1 (VCAM-1), and E-selectin, and of coagulation, including tissue factor and thrombomodulin, was assessed by real-time polymerase chain reaction. RESULTS Tofacitinib differentially inhibited production of IFNɣ, IL-17A, and IL-10 from PBMC cocultures with RA synoviocytes or ECs (all P < 0.001). In cocultures with ECs, tofacitinib reduced further IL-6 and IL-8 production (both P < 0.001). In ECs activated by TNF/IL-17A or indirectly via contact with activated PBMCs, tofacitinib decreased IL-6 upregulation but not that of IL-8, E-selectin, or tissue factor. Thrombomodulin was significantly decreased. VCAM-1 was greatly induced with a higher dose of tofacitinib in ECs incubated directly with added inflammatory cytokines (P < 0.05) or released by interaction with activated PBMCs (P < 0.001). CONCLUSION Tofacitinib inhibits synovium and vascular inflammation but fails to prevent the prothrombotic effects of inflammatory cytokines on ECs.
Collapse
Affiliation(s)
- Aliki Zavoriti
- Hôpital Edouard HerriotHospices Civils de LyonLyonFrance
| | - Pierre Miossec
- Hôpital Edouard HerriotHospices Civils de LyonLyonFrance
| |
Collapse
|
21
|
Zhang J, Hu F, Zhang J, Xie J, Wang Z, Lv L, Liang H, Liu Q, Chen R, Li H, Su W, Yan R, Chen Z, Wang Z, Tang H, Chang YN, Li J, Chen J, Shen M, Xing G, Chen K. Physical-Matched Nanoplatelets Boost Heterogeneous Thrombi Targeting Through Self-Adaptive Deformation for Thrombolysis and Endothelial Repairing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406262. [PMID: 39428893 DOI: 10.1002/smll.202406262] [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: 07/24/2024] [Revised: 09/14/2024] [Indexed: 10/22/2024]
Abstract
The heterogeneity of thrombi in terms of composition, structure, and blood rheology parameters presents a challenge for effective thrombus-targeting drug delivery. To address this, a self-adaptive nano-delivery system, termed D-PLT, is developed. It consists of platelet membrane-cloaked deformable mesoporous organic silicon dioxide nanocomposite, enabling it to respond to the challenge of the heterogeneity of thrombi in arteries and veins. The system exhibits progressive targeting, with the ability to target arterial and venous thrombosis and damaged blood vessels. D-PLT physically matches the pore structure of the thrombus by undergoing varied deformation, leading to advanced targeting and enrichment of arterial and venous thrombus. When co-loaded with the thrombolytic drug urokinase (UK) and the endothelium-protecting agent atorvastatin calcium (AT), the system improves rapid vascular opening of arterial and venous thrombosis in 90 min and provides up to 7 days of durable thrombolysis and recovery from endothelial dysfunction in vivo. This self-adaptive delivery system offers a promising strategy to overcome thrombus heterogeneity.
Collapse
Affiliation(s)
- Junhui Zhang
- Department of Biochemistry and Molecular Biology, Yanbian University Medical College, Yanji, Jilin, 133002, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Fan Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jing Xie
- State Key Laboratory of Explosion Science and Safety Protection, Institute of Technology Beijing, Beijing, 100081, P. R. China
| | - Zhiyu Wang
- State Key Laboratory of Explosion Science and Safety Protection, Institute of Technology Beijing, Beijing, 100081, P. R. China
| | - Linwen Lv
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Haojun Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiuyang Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Ranran Chen
- Department of Biochemistry and Molecular Biology, Yanbian University Medical College, Yanji, Jilin, 133002, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Wenxi Su
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Ruyu Yan
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Ziteng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Zhijie Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Hongyu Tang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Ya-Nan Chang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Juan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Jun Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Minghua Shen
- Department of Biochemistry and Molecular Biology, Yanbian University Medical College, Yanji, Jilin, 133002, China
| | - Gengmei Xing
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Kui Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| |
Collapse
|
22
|
Zheng B, Chen J, Xu Y, Wu W, Zhu Y, Cai W, Lin W, Shi C. Poly (β-amino esters)/Mobil Composition of Matter 41-mediated delivery of siIL-1β alleviates deep vein thrombosis in rat hind limbs. J Biomater Appl 2025; 39:648-660. [PMID: 39213651 DOI: 10.1177/08853282241280376] [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: 09/04/2024]
Abstract
Introduction: Deep vein thrombosis (DVT) is a major cause of cardiovascular disease-related deaths worldwide and is considered a thrombotic inflammatory disorder. IL-1β, as a key promoter of venous thrombus inflammation, is a potential target for DVT treatment. Constructing a nanocarrier system for intracellular delivery of siIL-1β to silence IL-1β may be an effective strategy for alleviating DVT. Methods: ELISA was used to detect the expression levels of IL-1β and t-PA in the serum of DVT patients and healthy individuals. In vitro, HUVEC cells were treated with IL-1β, and changes in VWF and t-PA expression levels were assessed. PBAE/MCM-41@siIL-1β (PM@siIL-1β) nano-complexes were synthesized, the characterization and biocompatibility of PM@siIL-1β were evaluated. A rat hind limb DVT model was established, and PM@siIL-1β was used to treat DVT rats. Morphology of the inferior vena cava, endothelial cell count, IL-1β, vWF, and t-PA levels, as well as changes in the p38 MAPK and NF-κB pathways, were examined in the different groups. Results: IL-1β and t-PA were highly expressed in DVT patients, and IL-1β treatment induced a decrease in VWF levels and an increase in t-PA levels in HUVEC cells. The synthesized PM@siIL-1β exhibited spherical shape, good stability, high encapsulation efficiency, and high drug loading capacity, with excellent biocompatibility. In the DVT model rats, the inferior vena cava was filled with blood clots, endothelial cells increased, IL-1β and VWF levels significantly increased, while t-PA levels were significantly downregulated. Treatment with PM@siIL-1β resulted in reduced thrombus formation, decreased endothelial cell count, and reversal of IL-1β, VWF, and t-PA levels. Furthermore, PM@siIL-1β treatment significantly inhibited p38 phosphorylation and upregulation of NF-κB expression in the DVT model group. Conclusion: IL-1β can be considered a therapeutic target for suppressing DVT inflammation. The synthesized PM@siIL-1β achieved efficient delivery and gene silencing of siIL-1β, demonstrating good therapeutic effects on rat hind limb DVT, including anti-thrombotic and anti-inflammatory effects, potentially mediated through the p38 MAPK and NF-κB pathways.
Collapse
Affiliation(s)
- Bingru Zheng
- Department of Interventional Vascular Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinjie Chen
- Department of Nephrology, Rui'an Third People's Hospital, Wenzhou, China
| | - Yizhou Xu
- Department of Interventional Vascular Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wanrui Wu
- Department of Interventional Vascular Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu Zhu
- Department of Interventional Vascular Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Cai
- Department of Interventional Vascular Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weili Lin
- Department of Ultrasound Imaging, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Changsheng Shi
- Department of Interventional Vascular Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
23
|
Ojaroodi AF, Jafarnezhad F, Eskandari Z, Keramat S, Stanek A. Recent Updates and Advances in the Association Between Vitamin D Deficiency and Risk of Thrombotic Disease. Nutrients 2024; 17:90. [PMID: 39796525 PMCID: PMC11722561 DOI: 10.3390/nu17010090] [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: 11/24/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Vitamin D (VD) is a vital lipophilic secosteroid hormone known for its essential role in maintaining skeletal health and regulating calcium and phosphate metabolism. Recent evidence has begun to illuminate its significance beyond bone health, particularly in relation to thrombosis-a condition characterized by blood clot formation within the vascular system that can lead to serious cardiovascular events such as myocardial infarction and stroke. VD deficiency, defined as a plasma 25-hydroxyVD level below 25 nmol/L, affects a substantial portion of the global population, with prevalence rates ranging from 8% to 18%. This study systematically explores the relationships between VD levels and the risk of thrombosis, investigating the underlying mechanisms including VD's anticoagulant properties, influence on inflammatory pathways, and interactions with endothelial cells. Epidemiological data suggest that low serum levels of VD correlate with an increased risk of venous thromboembolism (VTE), although the reported findings remain inconsistent. Mechanisms that potentially link VD to thrombotic risk include modulation of thrombomodulin and tissue factor expression, as well as enhancement of anti-inflammatory cytokines. Given the prevalence of VD insufficiency, particularly among populations with limited exposure to sunlight, this research highlights the urgent need for strategies to increase VD levels through dietary modifications and supplementation in order to prevent thrombotic events.
Collapse
Affiliation(s)
- Amirhossein Faghih Ojaroodi
- Hematology and Transfusion Medicine Ward, Department of Medical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran;
| | - Fatemeh Jafarnezhad
- Department of Hematology, Faculty of Medicine, Ferdowsi University of Mashhad, Mashhad 9177899191, Iran
| | - Zahra Eskandari
- Department of Hematology, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr 7518759577, Iran;
| | - Shayan Keramat
- VAS-European Independent Foundation in Angiology/Vascular Medicine, Via GB Grassi 74, 20157 Milan, Italy;
- Support Association of Patients of Buerger’s Disease, Buerger’s Disease NGO, Mashhad 9183785195, Iran
| | - Agata Stanek
- VAS-European Independent Foundation in Angiology/Vascular Medicine, Via GB Grassi 74, 20157 Milan, Italy;
- Department of Internal Medicine, Metabolic Diseases and Angiology, Faculty of Health Sciences in Katowice, Medical University of Silesia, Ziołowa 45/47, 40-635 Katowice, Poland
| |
Collapse
|
24
|
Weng C, Yu C, Yang GW, Jiang JS, Wu H. Association of red blood cell distribution width-albumin ratio with in-hospital mortality in abdominal aortic aneurysm patients. Medicine (Baltimore) 2024; 103:e40785. [PMID: 39654200 PMCID: PMC11630967 DOI: 10.1097/md.0000000000040785] [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: 10/07/2023] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024] Open
Abstract
To explore whether red blood cell distribution width-albumin ratio (RAR) is relevant to in-hospital mortality among abdominal aortic aneurysm (AAA). This is a retrospective study retrieving data from the MIMIC-IV database. Patients were divided into survivor or non-survivor groups by the in-hospital mortality. Receiver operating characteristic curve analysis, logistic regression models, subgroup analysis, interaction analysis, and restricted cubic spline analysis were conducted to analyze the correlation between RAR and in-hospital mortality. Then, we divided patients into 2 groups by an optimal cutoff value of RAR to identify the factors independently linked to RAR. Following this, the mediation analysis was conducted to reveal the potential regulatory path. Finally, we assessed the clinical value of RAR in secondary outcomes containing length of hospital stay, intensive care unit (ICU) admission, and ICU stay. Totally 770 participants with AAA were enrolled: 722 survivors and 48 non-survivors. Higher RAR was observed in the non-survivor group and its level performed satisfactorily in predicting in-hospital mortality. AAA patients were more likely to die during in-hospital with the increase of RAR (P < .05) and this linear correlation was revealed by restricted cubic spline (P non-linear > .05). Additionally, urea nitrogen and creatinine were independently related to RAR. RAR served as a mediator in the association of urea nitrogen/creatinine with in-hospital mortality. Finally, the length of hospital stay and ICU stay were longer in the RAR ≥ 4.658 group (P < .05). RAR is a potential risk predictor for in-hospital mortality in AAA patients. Further, RAR upregulation was significantly correlated with prolonged length of hospital stay and ICU stay.
Collapse
Affiliation(s)
- Chao Weng
- Department of Vascular Surgery, General Surgery, Cancer Center, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Cong Yu
- Department of Vascular Surgery, General Surgery, Cancer Center, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Guang-Wei Yang
- Department of Vascular Surgery, General Surgery, Cancer Center, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jin-Song Jiang
- Department of Vascular Surgery, General Surgery, Cancer Center, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hao Wu
- Department of Vascular Surgery, General Surgery, Cancer Center, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Liu Y, Huang T, Yap NA, Lim K, Ju LA. Harnessing the power of bioprinting for the development of next-generation models of thrombosis. Bioact Mater 2024; 42:328-344. [PMID: 39295733 PMCID: PMC11408160 DOI: 10.1016/j.bioactmat.2024.08.040] [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: 05/19/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Thrombosis, a leading cause of cardiovascular morbidity and mortality, involves the formation of blood clots within blood vessels. Current animal models and in vitro systems have limitations in recapitulating the complex human vasculature and hemodynamic conditions, limiting the research in understanding the mechanisms of thrombosis. Bioprinting has emerged as a promising approach to construct biomimetic vascular models that closely mimic the structural and mechanical properties of native blood vessels. This review discusses the key considerations for designing bioprinted vascular conduits for thrombosis studies, including the incorporation of key structural, biochemical and mechanical features, the selection of appropriate biomaterials and cell sources, and the challenges and future directions in the field. The advancements in bioprinting techniques, such as multi-material bioprinting and microfluidic integration, have enabled the development of physiologically relevant models of thrombosis. The future of bioprinted models of thrombosis lies in the integration of patient-specific data, real-time monitoring technologies, and advanced microfluidic platforms, paving the way for personalized medicine and targeted interventions. As the field of bioprinting continues to evolve, these advanced vascular models are expected to play an increasingly important role in unraveling the complexities of thrombosis and improving patient outcomes. The continued advancements in bioprinting technologies and the collaboration between researchers from various disciplines hold great promise for revolutionizing the field of thrombosis research.
Collapse
Affiliation(s)
- Yanyan Liu
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Tao Huang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Nicole Alexis Yap
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Khoon Lim
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- School of Medical Sciences, The University of Sydney, Darlington, NSW 2008, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW 2042, Australia
| |
Collapse
|
26
|
Yadav SN, Al Hasan MS, Das B, Shadin M, Rakib IH, Rohan F, Ansari SA, Ansari IA, Bhuia MS, Lima MA, Domiciano CB, Coutinho HDM, Islam MT. Assessment of clot-lysing and membrane-stabilizing capacity of ascorbic acid: In vitro approach with molecular docking. Toxicol Rep 2024; 13:101831. [PMID: 39687679 PMCID: PMC11646741 DOI: 10.1016/j.toxrep.2024.101831] [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: 10/15/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
This study aimed to evaluate the clot-lysing and membrane stabilizing capacities of ascorbic acid (AA) using in vitro and in silico methods. For this, we used in vitro clot lysis and hemolyzing tests to check the anti-atherothrombosis and membrane-stabilizing properties of AA, respectively. Additionally, molecular docking studies were performed to investigate AA's interactions with cyclooxygenase-1 (COX-1) and plasminogen enzymes. Findings suggest that AA exhibited a concentration-dependent effect, with 43.95 ± 1.27 % clot lysis and 64.46 ± 0.01 % membrane stabilization at 100 µg/mL. The IC50 values for clot lysis and membrane stabilization were 215.19 ± 1.09 and 57.21 ± 2.11 µg/mL, respectively. In silico analysis showed strong binding affinities of AA with COX-1 (-6.2 kcal/mol) and plasminogen (-5.8 kcal/mol), supporting its observed clot lysis and membrane protection activities. Taken together, AA showed moderate clot-lysing and robust membrane-stabilizing effects, which may be due to its strong antioxidant and anti-inflammatory properties. AA might be a good therapeutic agent for atherothrombosis and membrane damage, highlighting the need for further investigation into its underlying molecular mechanisms and potential clinical applications. AA shows promising clot-lysing and membrane-stabilizing effects, highlighting its therapeutic potential for atherothrombosis and membrane damage.
Collapse
Affiliation(s)
- Shuv Narayan Yadav
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Md. Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj 8100, Bangladesh
| | - Balaram Das
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Md. Shadin
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj 8100, Bangladesh
| | - Imam Hossen Rakib
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj 8100, Bangladesh
| | - Fazley Rohan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Irfan Aamer Ansari
- Department of Drug Science and Technology, University of Turin, Turin 10124, Italy
| | - Md. Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj 8100, Bangladesh
| | | | | | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj 8100, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna 9208, Bangladesh
| |
Collapse
|
27
|
Matsumoto H, Annen S, Mukai N, Ohshita M, Ogawa S, Okita M, Tanabe T, Takezawa M, Nakabayashi Y, Kikuchi S, Takeba J, Sato N. Association of endotheliopathy with coagulofibrinolytic reactions and disseminated intravascular coagulation after trauma: a retrospective observational study. Sci Rep 2024; 14:29630. [PMID: 39609563 PMCID: PMC11604942 DOI: 10.1038/s41598-024-81123-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024] Open
Abstract
We carried out a retrospective observational investigation to explore the association of endotheliopathy with coagulofibrinolytic reactions and the progression of disseminated intravascular coagulation (DIC) in adult trauma patients. We measured syndecan-1 (SDC-1), an indicator of endotheliopathy, and biomarkers of coagulofibrinolysis in 100 trauma patients immediately transferred to Ehime University Hospital. We evaluated the correlations between the coagulofibrinolytic parameters and SDC-1. We also investigated the association between SDC-1 elevations and the development of DIC, and determined the discriminators of DIC development. The median SDC-1 concentration was 82.7 (43.5-178.1) ng/mL. DIC developed in 16 patients (16.0%), and SDC-1 concentrations were significantly higher in DIC patients than in non-DIC patients (218.8 [134.5-798.2] ng/mL vs. 67.2 [39.6-114.5] ng/mL, p < 0.001). Receiver operating characteristic curve analysis revealed that the circulating SDC-1 level effectively predicted the progression of DIC, with an area under the curve of 0.862 (95% confidence interval [CI], 0.789-0.936). The optimal cut-off value was determined to be 92.5 ng/mL, yielding a sensitivity of 100.0% and a specificity of 67.8% (p < 0.001). A simple logistic regression analysis showed that a circulating SDC-1 concentration of > 92.5 ng/mL was significantly correlated with DIC progression (odds ratio [OR], 31.67; 95%CI: 3.97-252.31, p = 0.001). Many coagulofibrinolytic parameters were significantly correlated with SDC-1. Estimating the discriminators of DIC development by the least absolute shrinkage and selection operator (LASSO) and elastic-net regression analysis identified markers of coagulofibrinolytic activation, such as thrombin-antithrombin complex (TAT) and tissue plasminogen activator (tPA). A multivariate logistic regression model using TAT, tPA, and SDC-1 demonstrated that TAT and tPA, but not SDC-1, were independent factors predicting the development of DIC (TAT per 10 µg/L: OR, 1.14, 95%CI: 1.05-1.24, p = 0.003; tPA per 100pg/mL: OR, 1.03, 95%CI: 1.01-1.05, p = 0.003; SDC-1 per 10ng/mL: OR, 1.00, 95%CI: 0.99-1.01, p = 0.973). Mediation analysis showed that SDC-1 elevation was predominantly associated with the development of DIC indirectly through the increase in TAT (proportion mediated = 96.1%, p < 0.001), while there was no significant indirect effect of SDC-1 elevation on the role of TAT elevation in DIC development was observed (p = 0.340). The primary pathogenesis of DIC in the acute phase of trauma is likely driven by coagulofibrinolytic activation. Endotheliopathy, as reflected by elevated circulating levels of SDC-1, is strongly associated with coagulofibrinolytic responses. Although endotheliopathy may contribute to the early development of DIC through coagulation activation, its role appears to be limited.
Collapse
Affiliation(s)
- Hironori Matsumoto
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan.
| | - Suguru Annen
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Naoki Mukai
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Muneaki Ohshita
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Shirou Ogawa
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Mitsuo Okita
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Tsunenori Tanabe
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Mitsuaki Takezawa
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Yuki Nakabayashi
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Satoshi Kikuchi
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Jun Takeba
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Norio Sato
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| |
Collapse
|
28
|
Kikuchi Y, Wakabayashi N, Braghirolli DI, Pranke P, Kamiya H, Oyama K. Evaluation of the patency rate and endothelialization of a poly-ε-caprolactone, nanofiber sheet-based vascular graft using a rat abdominal aortic implantation model. Front Surg 2024; 11:1464155. [PMID: 39640198 PMCID: PMC11617548 DOI: 10.3389/fsurg.2024.1464155] [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: 07/13/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction The global increase in cardiovascular diseases has resulted in an augmented development of artificial small-caliber vascular grafts used in bypass graft surgeries, such as coronary and distal artery bypass graft surgeries. However, no consensus exists regarding the best method for creating vascular grafts. Poly-ε-caprolactone (PCL) is a biocompatible and biodegradable material that has been widely studied as a scaffold for tissue regeneration, inclusive of vascular grafts. In this study, a vascular graft was created from a PCL nanofiber sheet (PCL graft), and the performance thereof was examined using a rat abdominal aortic implantation model. Methods The PCL nanofiber sheets were created using an electrospinning machine. These nanofiber sheets were rolled up. Glue was applied between layers using a PCL solution to create a PCL nanofiber vascular graft, with an inner diameter of 1 mm. PCL grafts with 7 mm length were implanted into the abdominal aorta of rats. Thereafter, the patency was determined by pulsating blood flow from the hemiresection site of the distal aorta of the graft anastomosis, and endothelialization was examined using hematoxylin and eosin and immunofluorescent staining methods. Results The patency rate of the PCL graft at 2 weeks was 57.1% (12 of 21 cases), which is not satisfactory as a small-caliber vascular graft. Patent cases, however, revealed a CD31-positive endothelial cell layer in the inner lumen and autologous cell infiltration into the scaffold, indicating autologous vessel-like regeneration. By contrast, the occluded cases showed disassembly of the nanofiber layers; and the inner layers folded into the middle of the lumen. This observation suggested that the disassembled inner layer of the PCL graft disturbed the blood flow and triggered occlusion. Conclusions PCL grafts can exhibit autologous vessel-like regeneration; nonetheless, regarding patency, grafts made from rolled-up PCL nanofiber sheets have structural weaknesses. Further improvements are required to achieve a long-term and high patency rate for PCL grafts.
Collapse
Affiliation(s)
- Yuta Kikuchi
- Department of Cardiac Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Naohiro Wakabayashi
- Sapporo Cardiovascular Clinic, Department of Cardiovascular Surgery, Sapporo, Japan
| | - Daikelly I. Braghirolli
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Patricia Pranke
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Hiroyuki Kamiya
- Department of Cardiac Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Kyohei Oyama
- Department of Cardiac Surgery, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
29
|
Daniele A, Lucas SJE, Rendeiro C. Variability of flow-mediated dilation across lower and upper limb conduit arteries. Eur J Appl Physiol 2024; 124:3265-3278. [PMID: 38878074 PMCID: PMC11519148 DOI: 10.1007/s00421-024-05517-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/23/2024] [Indexed: 10/30/2024]
Abstract
Endothelial dysfunction is an early predictor of atherosclerosis and cardiovascular disease. Flow-mediated dilation (FMD) is the gold standard to assess endothelial function in humans. FMD reproducibility has been mainly assessed in the brachial artery (BA) with limited research in lower limb arteries. The purpose of this study was to compare FMD reproducibility in the upper limb BA and lower limb superficial femoral artery (SFA) in young healthy adults.Fifteen young healthy adults (nine males; six females) underwent FMD, resting diameter, velocity, and shear rate measurements on three occasions to determine intra-and inter-day reproducibility in both BA and SFA, assessed by coefficient of variation (CV), intraclass correlation coefficient (ICC), and Bland-Altman plots.BA FMD CVs (intra-day: 4.2%; inter-day: 8.7%) and ICCs (intra-day: 0.967; inter-day: 0.903) indicated excellent reproducibility and reliability, while for SFA FMD, both CVs (intra-day: 11.6%; inter-day: 26.7%) and ICCs (intra-day: 0.898; inter-day: 0.651) showed good/moderate reproducibility and reliability. BA FMD was significantly more reproducible than SFA FMD (p < 0.05). Diameter reproducibility was excellent and similar between arteries, while resting velocity and shear rate have lower reproducibility in the BA compared to SFA. Bland-Altman plots displayed no proportional and fixed bias between measurements.In summary, SFA FMD is less reproducible than BA FMD, with identical volume of ultrasound training. Given the increasing interest in using SFA FMD to test the efficacy of interventions targeting lower limb's vascular health and as a potential biomarker for peripheral arterial disease risk, future studies should ensure higher levels of training for adequate reproducibility.
Collapse
Affiliation(s)
- Alessio Daniele
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Samuel J E Lucas
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Catarina Rendeiro
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK.
| |
Collapse
|
30
|
Ahmaditabar P, Mahmoodi M, Taheri RA, Asefnejad A. Preparation and in vitro evaluation of tissue plasminogen activator-loaded nanoliposomes with anticoagulant coating. Biochim Biophys Acta Gen Subj 2024; 1868:130704. [PMID: 39178920 DOI: 10.1016/j.bbagen.2024.130704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/06/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024]
Abstract
The clinical efficacy of tissue plasminogen activator (tPA) is limited by its lack of specific delivery, requiring large therapeutic doses that increase the risk of intracerebral hemorrhage, bleeding at the surgical site, and patient mortality after angioplasty. To address these limitations, this study aimed to develop a chitosan polysulfate (CsPs)-coated liposomal formulation for the sustained release of tPA. The CsPs-coated liposomes containing tPA (Liposome-tPA/CsPs) were fabricated using the thin-film hydration technique and their properties were compared to tPA-encapsulated nanoliposomes without a coating layer (Liposome-tPA). Liposome-tPA/CsPs showed a quasi-spherical morphology with a hydrodynamic diameter of 110 nm, while Liposome-tPA had a diameter of 80 nm. The thermal analysis showed that the degradation temperature and glass transition temperature (Tg) of Liposome-tPA/CsPs were higher than that of tPA alone, indicating improved temperature stability. The in vitro release study demonstrated a slow and sustained release of tPA from the Liposome-tPA/CsPs, with a concentration of 0.02 mg/ml at 1 h and 0.23 mg/ml at 180 h. The CsPs coating layer enhanced the antibacterial and antioxidant activity of the nanoliposomes. Liposome-tPA/CsPs exhibited higher cell viability compared to Liposome-tPA. It also achieved a higher percentage of thrombolysis, with complete clot dissolution observed after 3 h of treatment. These findings suggest that the Liposome-tPA/CsPs can be a promising approach to overcome the limitations associated with the systemic administration of tPA, potentially enhancing its clinical efficacy while reducing the risk of adverse events.
Collapse
Affiliation(s)
- Parvin Ahmaditabar
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mahboobeh Mahmoodi
- Department of Biomedical Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran; Joint Reconstruction Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ramezan Ali Taheri
- Department of Biology, Faculty of Sciences, University of Tehran, Tehran, Iran
| | - Azadeh Asefnejad
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
31
|
Vogl BJ, Vitale E, Ahn S, Sularz A, Chavez Ponce A, Lo Russo GV, Collins J, Bavo AM, El Shaer A, Kramer A, Jia Y, Lulic D, De Beule M, Nielsen-Kudsk JE, De Backer O, Alkhouli M, Hatoum H. Flow Dynamic Factors Correlated With Device-Related Thrombosis After Left Atrial Appendage Occlusion. JACC. ADVANCES 2024; 3:101339. [PMID: 39493311 PMCID: PMC11530902 DOI: 10.1016/j.jacadv.2024.101339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 11/05/2024]
Abstract
Background Device-related thrombosis (DRT) occurs in up to 4% of patients undergoing left atrial appendage occlusion (LAAO) and is associated with substantial morbidity and mortality. However, its pathophysiology, predictors, and optimal management remain unclear. Objectives This study aims to assess flow dynamic factors correlating to DRT. Methods A multicenter registry of patients who underwent LAAO and had pre- and post-computed tomography imaging was used. Patient-specific 3-dimensional digital models of the left atrium were created, and finite element simulations were performed to implant an LAAO device into each model in a position that matched the clinical deployment. Computational fluid dynamic simulations were performed to quantify the following flow dynamic parameters: time averaged wall shear stress, oscillatory shear index, and endothelial cell activation potential. Results A total of 38 patients (19 with DRT and 19 without DRT) were included. Left atrium volumes and mitral valve areas were larger in the DRT cohort compared with controls. Patients with DRT had a significantly lower time averaged wall shear stress (1.76 ± 1.24 Pa vs 2.90 ± 2.70 Pa), a higher oscillatory shear index (0.19 ± 0.11 vs 0.17 ± 0.11), and a higher endothelial cell activation potential (0.23 ± 0.58 Pa-1 vs 0.17 ± 0.30 Pa-1) than the controls (P < 0.001 for all). Thrombus locations identified from in-vivo images correlated well with the flow dynamic parameters tested. Conclusions Flow dynamic parameters may be able to predict the risk of DRT after LAAO. Further investigation with a larger patient cohort and long-term follow-up is needed to assess the role of computational fluid dynamics in the risk stratification of patients considered for LAAO.
Collapse
Affiliation(s)
- Brennan J. Vogl
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, USA
| | - Emily Vitale
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, USA
| | - Sunyoung Ahn
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, USA
| | - Agata Sularz
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Gerardo V. Lo Russo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jeremy Collins
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ahmed El Shaer
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Anders Kramer
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Yuheng Jia
- Department of Cardiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Davorka Lulic
- Department of Cardiology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Ole De Backer
- Department of Cardiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mohamad Alkhouli
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Hoda Hatoum
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, USA
- Health Research Institute, Center of Biocomputing and Digital Health and Institute of Computing and Cybersystems, Michigan Technological University, Houghton, Michigan, USA
| |
Collapse
|
32
|
Avdonin PP, Blinova MS, Serkova AA, Komleva LA, Avdonin PV. Immunity and Coagulation in COVID-19. Int J Mol Sci 2024; 25:11267. [PMID: 39457048 PMCID: PMC11508857 DOI: 10.3390/ijms252011267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Discovered in late 2019, the SARS-CoV-2 coronavirus has caused the largest pandemic of the 21st century, claiming more than seven million lives. In most cases, the COVID-19 disease caused by the SARS-CoV-2 virus is relatively mild and affects only the upper respiratory tract; it most often manifests itself with fever, chills, cough, and sore throat, but also has less-common mild symptoms. In most cases, patients do not require hospitalization, and fully recover. However, in some cases, infection with the SARS-CoV-2 virus leads to the development of a severe form of COVID-19, which is characterized by the development of life-threatening complications affecting not only the lungs, but also other organs and systems. In particular, various forms of thrombotic complications are common among patients with a severe form of COVID-19. The mechanisms for the development of thrombotic complications in COVID-19 remain unclear. Accumulated data indicate that the pathogenesis of severe COVID-19 is based on disruptions in the functioning of various innate immune systems. The key role in the primary response to a viral infection is assigned to two systems. These are the pattern recognition receptors, primarily members of the toll-like receptor (TLR) family, and the complement system. Both systems are the first to engage in the fight against the virus and launch a whole range of mechanisms aimed at its rapid elimination. Normally, their joint activity leads to the destruction of the pathogen and recovery. However, disruptions in the functioning of these innate immune systems in COVID-19 can cause the development of an excessive inflammatory response that is dangerous for the body. In turn, excessive inflammation entails activation of and damage to the vascular endothelium, as well as the development of the hypercoagulable state observed in patients seriously ill with COVID-19. Activation of the endothelium and hypercoagulation lead to the development of thrombosis and, as a result, damage to organs and tissues. Immune-mediated thrombotic complications are termed "immunothrombosis". In this review, we discuss in detail the features of immunothrombosis associated with SARS-CoV-2 infection and its potential underlying mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Pavel V. Avdonin
- Koltzov Institute of Developmental Biology RAS, ul. Vavilova, 26, 119334 Moscow, Russia; (P.P.A.)
| |
Collapse
|
33
|
Duan L, Ma Y, Reisch B, Hadrovic E, Mach P, Kimmig R, Jahn M, Köninger A, Iannaccone A, Gellhaus A. Alteration in sB7-H4 Serum Levels and Placental Biomarker Expression after Therapeutic Plasma Exchange in Early-Onset Preeclampsia Patients. Int J Mol Sci 2024; 25:11082. [PMID: 39456867 PMCID: PMC11507903 DOI: 10.3390/ijms252011082] [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: 09/17/2024] [Revised: 10/12/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Therapeutic plasma exchange (TPE) is a widely used treatment for numerous diseases including pregnancy-related conditions. Our prior study on 20 early-onset preeclampsia patients undergoing TPE revealed a significant extension in pregnancy duration and reduced serum levels of sFlt-1, sFlt-1/PlGF, and sEndoglin. Here, we investigated the impact of TPE on serum sB7-H4, an immunological checkpoint molecule, and placental proteins (Flt-1, Eng, B7-H4, iNOS, TNF-α) in TPE-treated early-onset preeclampsia patients (N = 12, 23 + 2-28 + 5 weeks), conventionally treated counterparts (N = 12, 23 + 5-30 weeks), and gestational age-matched controls (N = 8, 22 + 4-31 + 6 weeks). Immunoblotting, ELISA, and co-immunohistochemistry were used for biomarker analysis, including placental inflammation factors (iNOS, TNF-α). The results showed that TPE extended pregnancy by a median of 6.5 days in this cohort of early-onset preeclampsia. Serum sB7-H4, sFlt-1, and sEndoglin levels decreased, along with reduced expression of their membrane-bound proteins in placental tissue upon TPE treatment. Moreover, TPE-treated patients displayed reduced placental inflammation compared to preeclampsia patients receiving standard-of-care treatment. In conclusion, TPE may improve pregnancy outcomes in early-onset preeclampsia by lowering circulating levels of sB7-H4, sFlt-1, and sEndoglin, as well as reducing placental inflammation. This translational approach holds promise for enhancing placental function and extending gestation in high-risk pregnancies including very preterm PE or HELLP cases.
Collapse
Affiliation(s)
- Liyan Duan
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, 45147 Essen, Germany; (L.D.); (Y.M.); (B.R.); (E.H.); (P.M.); (R.K.); (A.K.)
| | - Yuyang Ma
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, 45147 Essen, Germany; (L.D.); (Y.M.); (B.R.); (E.H.); (P.M.); (R.K.); (A.K.)
| | - Beatrix Reisch
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, 45147 Essen, Germany; (L.D.); (Y.M.); (B.R.); (E.H.); (P.M.); (R.K.); (A.K.)
| | - Elina Hadrovic
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, 45147 Essen, Germany; (L.D.); (Y.M.); (B.R.); (E.H.); (P.M.); (R.K.); (A.K.)
| | - Pawel Mach
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, 45147 Essen, Germany; (L.D.); (Y.M.); (B.R.); (E.H.); (P.M.); (R.K.); (A.K.)
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, 45147 Essen, Germany; (L.D.); (Y.M.); (B.R.); (E.H.); (P.M.); (R.K.); (A.K.)
| | - Michael Jahn
- Department of Nephrology, University of Duisburg-Essen, University Hospital Essen, 45147 Essen, Germany;
| | - Angela Köninger
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, 45147 Essen, Germany; (L.D.); (Y.M.); (B.R.); (E.H.); (P.M.); (R.K.); (A.K.)
- Department of Gynecology and Obstetrics, University Clinic St. Hedwig of the Order of St. John, 93049 Regensburg, Germany
| | - Antonella Iannaccone
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, 45147 Essen, Germany; (L.D.); (Y.M.); (B.R.); (E.H.); (P.M.); (R.K.); (A.K.)
| | - Alexandra Gellhaus
- Department of Gynecology and Obstetrics, University of Duisburg-Essen, 45147 Essen, Germany; (L.D.); (Y.M.); (B.R.); (E.H.); (P.M.); (R.K.); (A.K.)
| |
Collapse
|
34
|
Lira AL, Kohs TC, Moellmer SA, Shatzel JJ, McCarty OJ, Puy C. Substrates, Cofactors, and Cellular Targets of Coagulation Factor XIa. Semin Thromb Hemost 2024; 50:962-969. [PMID: 36940715 PMCID: PMC11069399 DOI: 10.1055/s-0043-1764469] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023]
Abstract
Coagulation factor XI (FXI) has increasingly been shown to play an integral role in several physiologic and pathological processes. FXI is among several zymogens within the blood coagulation cascade that are activated by proteolytic cleavage, with FXI converting to the active serine protease form (FXIa). The evolutionary origins of FXI trace back to duplication of the gene that transcribes plasma prekallikrein, a key factor in the plasma kallikrein-kinin system, before further genetic divergence led to FXI playing a unique role in blood coagulation. While FXIa is canonically known for activating the intrinsic pathway of coagulation by catalyzing the conversion of FIX into FIXa, it is promiscuous in nature and has been shown to contribute to thrombin generation independent of FIX. In addition to its role in the intrinsic pathway of coagulation, FXI also interacts with platelets, endothelial cells, and mediates the inflammatory response through activation of FXII and cleavage of high-molecular-weight kininogen to generate bradykinin. In this manuscript, we critically review the current body of knowledge surrounding how FXI navigates the interplay of hemostasis, inflammatory processes, and the immune response and highlight future avenues for research. As FXI continues to be clinically explored as a druggable therapeutic target, understanding how this coagulation factor fits into physiological and disease mechanisms becomes increasingly important.
Collapse
Affiliation(s)
- André L. Lira
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Tia C.L. Kohs
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Samantha A. Moellmer
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Joseph J. Shatzel
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Owen J.T. McCarty
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Cristina Puy
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
35
|
Oliveira-Paula GH, Martins AC, Ferrer B, Tinkov AA, Skalny AV, Aschner M. The impact of manganese on vascular endothelium. Toxicol Res 2024; 40:501-517. [PMID: 39345740 PMCID: PMC11436708 DOI: 10.1007/s43188-024-00260-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/10/2024] [Accepted: 07/26/2024] [Indexed: 10/01/2024] Open
Abstract
Manganese (Mn) is an essential trace element involved in various physiological processes, but excessive exposure may lead to toxicity. The vascular endothelium, a monolayer of endothelial cells within blood vessels, is a primary target of Mn toxicity. This review provides a comprehensive overview of the impact of Mn on vascular endothelium, focusing on both peripheral and brain endothelial cells. In vitro studies have demonstrated that high concentrations of Mn can induce endothelial cell cytotoxicity, increase permeability, and disrupt cell-cell junctions through mechanisms involving oxidative stress, mitochondrial damage, and activation of signaling pathways, such as Smad2/3-Snail. Conversely, low concentrations of Mn may protect endothelial cells from the deleterious effects of high glucose and advanced glycation end-products. In the central nervous system, Mn can cross the blood-brain barrier (BBB) and accumulate in the brain parenchyma, leading to neurotoxicity. Several transport mechanisms, including ZIP8, ZIP14, and SPCA1, have been identified for Mn uptake by brain endothelial cells. Mn exposure can impair BBB integrity by disrupting tight junctions and increasing permeability. In vivo studies have corroborated these findings, highlighting the importance of endothelial barriers in mediating Mn toxicity in the brain and kidneys. Maintaining optimal Mn homeostasis is crucial for preserving endothelial function, and further research is needed to develop targeted therapeutic strategies to prevent or mitigate the adverse effects of Mn overexposure. Graphical Abstract
Collapse
Affiliation(s)
| | - Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - Beatriz Ferrer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - Alexey A. Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003 Russia
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435 Russia
| | - Anatoly V. Skalny
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003 Russia
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435 Russia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| |
Collapse
|
36
|
Augustin HG, Koh GY. A systems view of the vascular endothelium in health and disease. Cell 2024; 187:4833-4858. [PMID: 39241746 DOI: 10.1016/j.cell.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 09/09/2024]
Abstract
The dysfunction of blood-vessel-lining endothelial cells is a major cause of mortality. Although endothelial cells, being present in all organs as a single-cell layer, are often conceived as a rather inert cell population, the vascular endothelium as a whole should be considered a highly dynamic and interactive systemically disseminated organ. We present here a holistic view of the field of vascular research and review the diverse functions of blood-vessel-lining endothelial cells during the life cycle of the vasculature, namely responsive and relaying functions of the vascular endothelium and the responsive roles as instructive gatekeepers of organ function. Emerging translational perspectives in regenerative medicine, preventive medicine, and aging research are developed. Collectively, this review is aimed at promoting disciplinary coherence in the field of angioscience for a broader appreciation of the importance of the vasculature for organ function, systemic health, and healthy aging.
Collapse
Affiliation(s)
- Hellmut G Augustin
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ), 69120 Heidelberg, Germany.
| | - Gou Young Koh
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea; Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
37
|
Bryson C, Lodha C, Miller S. Multifocal Infarction Along the Alimentary Canal in the Context of Ostensible Salmonellosis: A Case Report. Cureus 2024; 16:e69707. [PMID: 39429332 PMCID: PMC11490280 DOI: 10.7759/cureus.69707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
Ischemic and/or infarction events of the alimentary canal are uncommon but potentially disastrous injuries of the digestive system that often portend a poor prognosis. Alimentary ischemia occurs when the vascular supply to one of the component conduit organs is disrupted or blocked, resulting in decreased tissue perfusion, subsequent necrosis, perforation, and even death if proper perfusion is not restored. We report a case here of a 67-year-old female who originally presented to the emergency department (ED) with nausea, vomiting, diarrhea, and progressively worsening abdominal pain. Conservative therapies that were initially employed failed to provide lasting symptom relief, and the patient was admitted for a more in-depth diagnostic workup and closer monitoring. During subsequent days of her resulting hospital stay, the patient had a positive result for Salmonella spp. on a stool PCR assay, an increasing leukocytosis, and the presence of several other worrisome laboratory abnormalities. Despite appropriate antibiotics and aggressive fluid resuscitation efforts, the patient's abdominal pain and laboratory profile continued to progressively worsen. At one point, the patient's condition perilously worsened, necessitating an emergent exploratory laparotomy. During the course of this surgery and subsequent surgeries, the patient was found to have multiple areas of infarction present including at her esophagus, stomach, duodenum, proximal jejunum, and right colon. Additionally, evidence of a metastatic neuroendocrine tumor of gastrointestinal (GI) origin was also incidentally found. Several subsequent surgical operations were required to repair the extensive tissue damage that the patient had sustained, and the patient's resulting hospital stay was complicated repeatedly by several different secondary infections and surgical complications. Attempts to determine the underlying cause for the ischemic events this patient experienced failed to yield definitive results, and no evidence for any arterial insufficiency or emboli was ever discovered. Despite this, a review of the histopathologic and laboratory findings from the tissue resected from the patient did find information to suggest that a relatively localized but severe venous thrombotic process likely occurred in the patient's alimentary vasculature that directly led to her presentation. Venous thrombosis of the mesenteric vessels and in the other vascular planes of the alimentary canal is often insidious in its presentation and poses a unique diagnostic challenge to clinicians. This case is significant because it illustrates the diagnostic complexity and difficulty imposed by mesenteric ischemia, especially cases resulting from mesenteric venous thrombosis (MVT) due to their often more indolent and atypical presentation. In short, a high level of clinical suspicion and familiarity with this ailment and its risk factors should be maintained because, in the absence of timely intervention, significant morbidity and/or mortality are likely to result.
Collapse
Affiliation(s)
- Caleb Bryson
- Infectious Disease, Edward Via College of Osteopathic Medicine, Spartanburg, USA
| | - Chirag Lodha
- Internal Medicine, Edward Via College of Osteopathic Medicine, Spartanburg, USA
| | - Stanley Miller
- Internal Medicine, Edward Via College of Osteopathic Medicine, Spartanburg, USA
| |
Collapse
|
38
|
Iqbal SJ, Baloch ZQ, Malik J, Bhimani N, Mehmoodi A, Gupta V. Bridging With Low-Molecular-Weight Heparin Versus Antiplatelet Therapy in Patients Undergoing Noncardiac Surgery After Percutaneous Coronary Intervention: A Comprehensive Review. Clin Cardiol 2024; 47:e70008. [PMID: 39262104 PMCID: PMC11390793 DOI: 10.1002/clc.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND This review article discussed the use of bridging therapy with low-molecular-weight heparin (LMWH) in patients who undergo noncardiac surgery (NCS) after percutaneous coronary intervention (PCI). HYPOTHESES Patients who undergo PCI are at an increased risk of thrombotic events due to their underlying cardiovascular disease. However, many of these patients may require NCS at some point in their lives, which poses a significant challenge for clinicians as they balance the risk of thrombotic events against the risk of bleeding associated with antithrombotic therapy. RESULTS This review evaluates the current evidence on the use of bridging therapy with LMWH in patients undergoing NCS after PCI, focusing on outcomes related to the efficacy and safety of antithrombotic therapy. The article also discusses the limitations of the current evidence and highlights areas where further research is needed to optimize the management of antithrombotic therapy in this patient population. CONCLUSION The goal of this review was to provide clinicians with a comprehensive summary of the available evidence to guide clinical decision-making and improve patient outcomes.
Collapse
Affiliation(s)
- Syed Javaid Iqbal
- Department of Cardiovascular ResearchCardiovascular Analytics GroupIslamabadPakistan
| | | | - Jahanzeb Malik
- Department of Cardiovascular ResearchCardiovascular Analytics GroupIslamabadPakistan
| | - Nikeeta Bhimani
- Department of Cardiovascular ResearchCardiovascular Analytics GroupIslamabadPakistan
| | - Amin Mehmoodi
- Department of MedicineIbn e Seena HospitalKabulAfghanistan
| | - Vishal Gupta
- Department of Interventional CardiologyAscension Borgess HospitalMichiganUSA
| |
Collapse
|
39
|
Kunutsor SK, Kurl S, Laukkanen JA. Cardiorespiratory fitness, atrial fibrillation and stroke: a review of the evidence in 2024. Expert Rev Cardiovasc Ther 2024; 22:493-508. [PMID: 39329169 DOI: 10.1080/14779072.2024.2409440] [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: 04/19/2024] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
INTRODUCTION The body of evidence linking cardiorespiratory fitness (CRF) levels with the risk of atrial fibrillation (AF) and stroke - two interconnected cardiovascular conditions - is not entirely consistent. Furthermore, specific CRF thresholds beyond which the risk of AF or stroke might not decrease are not well defined. AREAS COVERED This review summarizes research evidence on the role of CRF in the development of AF and stroke including dose-response relationships in general population participants, explores the biological mechanisms through which CRF may exert its effects, assesses the potential implications for clinical care and population health, identifies gaps in the current evidence, and suggest directions for future research. MEDLINE and Embase were searched from inception until July 2024 to identify observational longitudinal and interventional studies as well as systematic reviews and meta-analyses related to these study designs. EXPERT OPINION In the general population, increasing levels of CRF, achieved through consistent physical activity, can significantly reduce the likelihood of developing AF and stroke. The findings also advocate for a tailored approach to exercise prescriptions, acknowledging the plateau in benefits for AF risk beyond certain CRF levels, while advocating for higher intensity or prolonged activity to further reduce stroke risk.
Collapse
Affiliation(s)
- Setor K Kunutsor
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, UK
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Saint Boniface Hospital, Winnipeg, Manitoba, Canada
| | - Sudhir Kurl
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Brain Research Unit, Department of Neurology, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jari A Laukkanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Wellbeing Services County of Central Finland, Jyväskylä, Finland
| |
Collapse
|
40
|
Groten SA, Smit ER, van den Biggelaar M, Hoogendijk AJ. The proteomic landscape of in vitro cultured endothelial cells across vascular beds. Commun Biol 2024; 7:989. [PMID: 39143368 PMCID: PMC11324761 DOI: 10.1038/s42003-024-06649-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 07/29/2024] [Indexed: 08/16/2024] Open
Abstract
Blood vessel endothelial cells (EC) display heterogeneity across vascular beds, which is anticipated to drive site-specific vascular pathology. This heterogeneity is assessed using transcriptomics in vivo, and functional assays in vitro, but how proteomes compare across human in vitro cultured ECs remains incompletely characterized. We generated an in-depth human EC proteomic landscape (>8000 proteins) across six organs and two in vitro models in steady-state and upon IFNγ-induced inflammation. EC proteomes displayed a high similarity and organ-specific proteins were limited. Variation between ECs was mainly based on proliferation and differentiation processes in which Blood outgrowth endothelial cells (BOEC) and Human umbilical vein cells (HUVEC) represented the extremes of proteomic phenotypes. The IFNγ response was highly conserved across all samples. Harnessing dynamics in protein abundances we delineated VWF and VE-Cadherin correlation networks. This EC landscape provides an extensive proteomic addition in studying EC biology and heterogeneity from an in vitro perspective.
Collapse
Affiliation(s)
- Stijn A Groten
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Eva R Smit
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | | | - Arie J Hoogendijk
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands.
| |
Collapse
|
41
|
Das D, Jothimani G, Banerjee A, Dey A, Duttaroy AK, Pathak S. A brief review on recent advances in diagnostic and therapeutic applications of extracellular vesicles in cardiovascular disease. Int J Biochem Cell Biol 2024; 173:106616. [PMID: 38992790 DOI: 10.1016/j.biocel.2024.106616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024]
Abstract
Extracellular vesicles (EVs) are important mediators of intercellular communication within the cardiovascular system, playing essential roles in physiological homeostasis and contributing to the pathogenesis of various cardiovascular diseases (CVDs). However, their potential as diagnostic biomarkers and therapeutic agents in rare cardiovascular diseases, such as valvular heart disease (VHD) and cardiomyopathies, remains largely unexplored. This review comprehensively emphasizes recent advancements in extracellular vesicle research, explicitly highlighting their growing significance in diagnosing and potentially treating rare cardiovascular diseases, with a particular focus on valvular heart disease and cardiomyopathies. We highlight the potential of extracellular vesicle-based liquid biopsies as non-invasive tools for early disease detection and risk stratification, showcasing specific extracellular vesicle-associated biomarkers (proteins, microRNAs, lipids) with diagnostic and prognostic value. Furthermore, we discussed the therapeutic promise of extracellular vesicles derived from various sources, including stem cells and engineered extracellular vesicles, for cardiac repair and regeneration through their ability to modulate inflammation, promote angiogenesis, and reduce fibrosis. By integrating the findings and addressing critical knowledge gaps, this review aims to stimulate further research and innovation in extracellular vesicle-based diagnostics and therapeutics of cardiovascular disease.
Collapse
Affiliation(s)
- Diptimayee Das
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Tamil Nadu 603103, India
| | - Ganesan Jothimani
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Tamil Nadu 603103, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Tamil Nadu 603103, India
| | - Amit Dey
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Tamil Nadu 603103, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, Norway.
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Tamil Nadu 603103, India.
| |
Collapse
|
42
|
Liu Z, Zhang X, Xiong S, Huang S, Ding X, Xu M, Yao J, Liu S, Zhao F. Endothelial dysfunction of syphilis: Pathogenesis. J Eur Acad Dermatol Venereol 2024; 38:1478-1490. [PMID: 38376088 DOI: 10.1111/jdv.19899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 01/19/2024] [Indexed: 02/21/2024]
Abstract
Treponema pallidum is the causative factor of syphilis, a sexually transmitted disease (STD) characterized by perivascular infiltration of inflammatory cells, vascular leakage, swelling and proliferation of endothelial cells (ECs). The endothelium lining blood and lymphatic vessels is a key barrier separating body fluids from host tissues and is a major target of T. pallidum. In this review, we focus on how T. pallidum establish intimate interactions with ECs, triggering endothelial dysfunction such as endothelial inflammation, abnormal repairment and damage of ECs. In addition, we summarize that migration and invasion of T. pallidum across vascular ECs may occur through two pathways. These two mechanisms of transendothelial migration are paracellular and cholesterol-dependent, respectively. Herein, clarifying the relationship between T. pallidum and endothelial dysfunction is of great significance to provide novel strategies for diagnosis and prevention of syphilis, and has a great potential prospect of clinical application.
Collapse
Affiliation(s)
- Zhaoping Liu
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Xiaohong Zhang
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Shun Xiong
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Shaobin Huang
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Xuan Ding
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Man Xu
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Jiangchen Yao
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Shuangquan Liu
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Feijun Zhao
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
43
|
Wang X, He B. Endothelial dysfunction: molecular mechanisms and clinical implications. MedComm (Beijing) 2024; 5:e651. [PMID: 39040847 PMCID: PMC11261813 DOI: 10.1002/mco2.651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 07/24/2024] Open
Abstract
Cardiovascular disease (CVD) and its complications are a leading cause of death worldwide. Endothelial dysfunction plays a crucial role in the initiation and progression of CVD, serving as a pivotal factor in the pathogenesis of cardiovascular, metabolic, and other related diseases. The regulation of endothelial dysfunction is influenced by various risk factors and intricate signaling pathways, which vary depending on the specific disease context. Despite numerous research efforts aimed at elucidating the mechanisms underlying endothelial dysfunction, the precise molecular pathways involved remain incompletely understood. This review elucidates recent research findings on the pathophysiological mechanisms involved in endothelial dysfunction, including nitric oxide availability, oxidative stress, and inflammation-mediated pathways. We also discuss the impact of endothelial dysfunction on various pathological conditions, including atherosclerosis, heart failure, diabetes, hypertension, chronic kidney disease, and neurodegenerative diseases. Furthermore, we summarize the traditional and novel potential biomarkers of endothelial dysfunction as well as pharmacological and nonpharmacological therapeutic strategies for endothelial protection and treatment for CVD and related complications. Consequently, this review is to improve understanding of emerging biomarkers and therapeutic approaches aimed at reducing the risk of developing CVD and associated complications, as well as mitigating endothelial dysfunction.
Collapse
Affiliation(s)
- Xia Wang
- Department of CardiologyShanghai Chest Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ben He
- Department of CardiologyShanghai Chest Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
44
|
Mueller TT, Pilartz M, Thakur M, LangHeinrich T, Luo J, Block R, Hoeflinger JKL, Meister S, Karaj F, Perez LG, Öllinger R, Engleitner T, Thoss J, Voelkl M, Tersteeg C, Koedel U, Kohlmaier AZ, Teupser D, Wygrecka M, Ye H, Preissner KT, Radbruch H, Elezkurtaj S, Mack M, Von Hundelshausen P, Weber C, Massberg S, Schulz C, Rad R, Huber S, Ishikawa-Ankerhold H, Engelmann B. Mutual regulation of CD4 + T cells and intravascular fibrin in infections. Haematologica 2024; 109:2487-2499. [PMID: 38572559 PMCID: PMC11290509 DOI: 10.3324/haematol.2023.284619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/25/2024] [Indexed: 04/05/2024] Open
Abstract
Innate myeloid cells especially neutrophils and their extracellular traps are known to promote intravascular coagulation and thrombosis formation in infections and various other conditions. Innate myeloid cell-dependent fibrin formation can support systemic immunity while its dysregulation enhances the severity of infectious diseases. Less is known about the immune mechanisms preventing dysregulation of fibrin homeostasis in infection. During experimental systemic infections local fibrin deposits in the liver microcirculation cause rapid arrest of CD4+ T cells. Arrested T-helper cells mostly represent Th17 cells that partially originate from the small intestine. Intravascular fibrin deposits activate mouse and human CD4+ T cells which can be mediated by direct fibrin-CD4+ T-cell interactions. Activated CD4+ T cells suppress fibrin deposition and microvascular thrombosis by directly counteracting coagulation activation by neutrophils and classical monocytes. T-cell activation, which is initially triggered by IL-12p40- and MHC-II-dependent mechanisms, enhances intravascular fibrinolysis via LFA-1. Moreover, CD4+ T cells disfavor the association of the thrombin-activatable fibrinolysis inhibitor (TAFI) with fibrin whereby fibrin deposition is increased by TAFI in the absence but not in the presence of T cells. In human infections thrombosis development is inversely related to microvascular levels of CD4+ T cells. Thus, fibrin promotes LFA-1-dependent T-helper cell activation in infections which drives a negative feedback cycle that rapidly restricts intravascular fibrin and thrombosis development.
Collapse
Affiliation(s)
- Tonina T Mueller
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich, Germany; Medizinische Klinik I, Klinikum der Universität München, LMU, Munich.
| | - Mona Pilartz
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Manovriti Thakur
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Torben LangHeinrich
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Junfu Luo
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Rebecca Block
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Jonathan K L Hoeflinger
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Sarah Meister
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Flavio Karaj
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Laura Garcia Perez
- 1. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| | - Rupert Öllinger
- Institut für Molekulare Onkologie und Funktionelle Genomik, Technische Universität München, Munich
| | - Thomas Engleitner
- Institut für Molekulare Onkologie und Funktionelle Genomik, Technische Universität München, Munich
| | - Jakob Thoss
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Michael Voelkl
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Claudia Tersteeg
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk
| | - Uwe Koedel
- Neurologische Klinik, Klinikum der Universität München,LMU, Munich
| | - Alexander Zigman Kohlmaier
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Daniel Teupser
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich
| | - Malgorzata Wygrecka
- Center for Infection and Genomics of the Lung (CIGL), Justus-Liebig-Universität, Giessen
| | - Haifeng Ye
- Institute of Regenerative Biology and Medicine, Helmholtz-Zentrum München, Munich
| | | | - Helena Radbruch
- Institut für Neuropathologie, Charité - Universitätsmedizin, Berlin
| | | | - Matthias Mack
- Medizinische Klinik II, University of Regensburg, Regensburg
| | - Philipp Von Hundelshausen
- Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten, Ludwig-Maximilians-Universität, Munich
| | - Christian Weber
- Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten, Ludwig-Maximilians-Universität, Munich
| | - Steffen Massberg
- Medizinische Klinik I, Klinikum der Universität München, LMU, Munich
| | - Christian Schulz
- Medizinische Klinik I, Klinikum der Universität München, LMU, Munich
| | - Roland Rad
- Institut für Molekulare Onkologie und Funktionelle Genomik, Technische Universität München, Munich
| | - Samuel Huber
- 1. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| | | | - Bernd Engelmann
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU), Munich.
| |
Collapse
|
45
|
Nunes M, Vlok M, Proal A, Kell DB, Pretorius E. Data-independent LC-MS/MS analysis of ME/CFS plasma reveals a dysregulated coagulation system, endothelial dysfunction, downregulation of complement machinery. Cardiovasc Diabetol 2024; 23:254. [PMID: 39014464 PMCID: PMC11253362 DOI: 10.1186/s12933-024-02315-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/16/2024] [Indexed: 07/18/2024] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating chronic condition that is characterized by unresolved fatigue, post-exertion symptom exacerbation (PESE), cognitive dysfunction, orthostatic intolerance, and other symptoms. ME/CFS lacks established clinical biomarkers and requires further elucidation of disease mechanisms. A growing number of studies demonstrate signs of hematological and cardiovascular pathology in ME/CFS cohorts, including hyperactivated platelets, endothelial dysfunction, vascular dysregulation, and anomalous clotting processes. To build on these findings, and to identify potential biomarkers that can be related to pathophysiology, we measured differences in protein expression in platelet-poor plasma (PPP) samples from 15 ME/CFS study participants and 10 controls not previously infected with SARS-CoV-2, using DIA LC-MS/MS. We identified 24 proteins that are significantly increased in the ME/CFS group compared to the controls, and 21 proteins that are significantly downregulated. Proteins related to clotting processes - thrombospondin-1 (important in platelet activation), platelet factor 4, and protein S - were differentially expressed in the ME/CFS group, suggestive of a dysregulated coagulation system and abnormal endothelial function. Complement machinery was also significantly downregulated, including C9 which forms part of the membrane attack complex. Additionally, we identified a significant upregulation of lactotransferrin, protein S100-A9, and an immunoglobulin variant. The findings from this experiment further implicate the coagulation and immune system in ME/CFS, and bring to attention the pathology of or imposed on the endothelium. This study highlights potential systems and proteins that require further research with regards to their contribution to the pathogenesis of ME/CFS, symptom manifestation, and biomarker potential, and also gives insight into the hematological and cardiovascular risk for ME/CFS individuals affected by diabetes mellitus.
Collapse
Affiliation(s)
- Massimo Nunes
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch, 7602, South Africa
| | - Mare Vlok
- Central Analytical Facility: Mass Spectrometry, Stellenbosch University, Tygerberg Campus, Room 6054, Clinical Building, Francie Van Zijl Drive Tygerberg, Cape Town, 7505, South Africa
| | - Amy Proal
- PolyBio Research Foundation, 7900 SE 28th ST, Suite 412, Mercer Island, DC, 98040, USA
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch, 7602, South Africa.
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool, L69 7ZB, UK.
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Chemitorvet 200, 2800, Kongens Lyngby, Denmark.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch, 7602, South Africa.
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool, L69 7ZB, UK.
| |
Collapse
|
46
|
Feng W, Qiao J, Tan Y, Liu Q, Wang Q, Yang B, Yang S, Cui L. Interaction of antiphospholipid antibodies with endothelial cells in antiphospholipid syndrome. Front Immunol 2024; 15:1361519. [PMID: 39044818 PMCID: PMC11263079 DOI: 10.3389/fimmu.2024.1361519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease with arteriovenous thrombosis and recurrent miscarriages as the main clinical manifestations. Due to the complexity of its mechanisms and the diversity of its manifestations, its diagnosis and treatment remain challenging issues. Antiphospholipid antibodies (aPL) not only serve as crucial "biomarkers" in diagnosing APS but also act as the "culprits" of the disease. Endothelial cells (ECs), as one of the core target cells of aPL, bridge the gap between the molecular level of these antibodies and the tissue and organ level of pathological changes. A more in-depth exploration of the relationship between ECs and the pathogenesis of APS holds the potential for significant advancements in the precise diagnosis, classification, and therapy of APS. Many researchers have highlighted the vital involvement of ECs in APS and the underlying mechanisms governing their functionality. Through extensive in vitro and in vivo experiments, they have identified multiple aPL receptors on the EC membrane and various intracellular pathways. This article furnishes a comprehensive overview and summary of these receptors and signaling pathways, offering prospective targets for APS therapy.
Collapse
Affiliation(s)
- Weimin Feng
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Yuan Tan
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Qi Liu
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| | - Qingchen Wang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Boxin Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
- Institute of Medical Technology, Health Science Centre, Peking University, Beijing, China
| |
Collapse
|
47
|
Cevik E, Albadawi H, Zhang Z, Demirlenk Y, Atar D, Keum C, Kim J, Graf E, Gunduz S, Rehman S, Oklu R. Catheter-Directed Ionic Liquid Embolic Agent for Rapid Portal Vein Embolization, Segmentectomy, and Bile Duct Ablation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402570. [PMID: 38678378 PMCID: PMC11257814 DOI: 10.1002/adma.202402570] [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: 02/19/2024] [Revised: 04/21/2024] [Indexed: 04/29/2024]
Abstract
Embolic materials currently in use for portal vein embolization (PVE) do not treat the tumor, which poses a risk for tumor progression during the interval between PVE and surgical resection. Here, is developed an ionic-liquid-based embolic material (LEAD) for portal vein embolization, liver ablation, and drug delivery. LEAD is optimized and characterized for diffusivity, X-ray visibility, and cytotoxicity. In the porcine renal embolization model, LEAD delivered from the main renal artery reached vasculature down to 10 microns with uniform tissue ablation and delivery of small and large therapeutics. In non-survival and survival porcine experiments, successful PVE is achieved in minutes, leading to the expected chemical segmentectomy, and delivery of a large protein drug (i.e., Nivolumab) with LEAD. In cholangiocarcinoma mouse tumor models and in ex vivo human tumors, LEAD consistently achieved an effective ablation and wide drug distribution. Furthermore, various strains of drug-resistant patient-derived bacteria showed significant susceptibility to LEAD, suggesting that LEAD may also prevent infectious complications resulting from tissue ablation. With its capabilities to embolize, ablate, and deliver therapeutics, ease of use, and a high safety profile demonstrated in animal studies, LEAD offers a potential alternative to tumor ablation with or without PVE for FLR growth.
Collapse
Affiliation(s)
- Enes Cevik
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Hassan Albadawi
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Zefu Zhang
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Yusuf Demirlenk
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Dila Atar
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Chris Keum
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Jinjoo Kim
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Erin Graf
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, Arizona 85054, USA
| | - Seyda Gunduz
- Department of Medical Oncology, Istinye University Bahcesehir Liv Hospital, Istanbul 34517, Turkey
| | - Suliman Rehman
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Rahmi Oklu
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| |
Collapse
|
48
|
Mhlongo G, Mnisi CM. Dietary incorporation of brown seaweed spent oyster mushroom substrate alters growth performance, physiological responses and meat quality parameters in Boschveld roosters. Sci Rep 2024; 14:14414. [PMID: 38909163 PMCID: PMC11193798 DOI: 10.1038/s41598-024-65338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 06/19/2024] [Indexed: 06/24/2024] Open
Abstract
Use of brown seaweed (Ecklonia maxima) as a nutraceutical source in indigenous chicken diets is limited by high dietary fibre levels. Inoculating seaweeds with oyster mushroom (Pleurotus ostreatus) spawn (OMS) could enhance the utility of the spent mushroom substrate (SMS). This study investigated the effect of feeding incremental levels of brown seaweed SMS on growth performance, physiological responses, and meat quality parameters in Boschveld roosters. A total of 324, 4-week-old Boschveld roosters were weighed and randomly allotted to 36 pens (9 birds per pen) to produce six replicates per dietary treatment. The diets were formulated as follows: a standard grower diet (CON); and CON containing 150 g/kg of brown seaweed inoculated with OMS at 0 (SMS0), 20 (SMS20), 30 (SMS30), 40 (SMS40) and 50% (SMS50). Birds fed diet CON had the least feed intake (p < 0.05) than all the other SMS treatment levels in weeks 7, 8, 12, 14 and 15. Diet SMS40 promoted higher (p < 0.05) body weight gain (BWG) than CON in weeks 6, 7, 9 and 14. Gain-to-feed ratio linearly increased in weeks 7 [R2 = 0.288; p = 0.010], 11 [R2 = 0.581, p = 0.0001] and 14 [R2 = 0.389, p = 0.004], respectively. Quadratic responses (p < 0.05) were observed for BWG in week 5, white blood cells, heterophils, platelets, lymphocytes, monocytes, and relative spleen and large intestine weights as OMS levels increased. Linear increases were recorded for slaughter [R2 = 0.197, p = 0.017] and breast weights [R2 = 0.197, p = 0.020] as OMS levels increased. Diet SMS0 promoted higher (p < 0.05) relative caeca weights than the CON and SMS treatment groups. Neither quadratic nor linear responses (p > 0.05) were observed for breast meat quality parameters. In conclusion, feeding brown seaweed SMS improved growth performance and slaughter weight, altered some blood parameters and internal organs, without affecting breast meat quality of Boschveld roosters. Based on the quadratic response for BWG, the optimum OMS level was deduced at 20% in a brown seaweed-based Boschveld rooster diet.
Collapse
Affiliation(s)
- Godfrey Mhlongo
- Department of Animal Science, School of Agricultural Science, North-West University, Private Bag x2046, Mafikeng, 2745, South Africa.
- Faculty of Agriculture and Natural Sciences, School of Agricultural Sciences, University of Mpumalanga, Mbombela, South Africa.
| | - Caven M Mnisi
- Department of Animal Science, School of Agricultural Science, North-West University, Private Bag x2046, Mafikeng, 2745, South Africa
- Food Security and Safety Focus Area, Faculty of Natural and Agricultural Science, North-West University, Mafikeng, South Africa
| |
Collapse
|
49
|
Nechita LC, Ignat MD, Balta AAS, Barbu RE, Baroiu L, Voinescu DC, Nechita A, Debita M, Busila C, Stefanopol IA. The Impact of Cardiovascular Antecedents on the Prognosis of COVID-19 Critically Ill Patients. J Clin Med 2024; 13:3518. [PMID: 38930047 PMCID: PMC11205074 DOI: 10.3390/jcm13123518] [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: 04/24/2024] [Revised: 05/17/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Background/Objectives: The objective of the study is to analyze the impact of cardiovascular history on mortality in COVID-19 patients, hospitalized in the intensive care unit with indications for continuous positive airway pressure (CPAP) and subsequently mechanical ventilation, without oncological disease. Methods: A retrospective observational study was carried out on a group of 108 critical COVID-19 patients. We compared demographic data, paraclinical and clinical parameters, days of hospitalization, and mortality rate between two groups of patients, one group with a history of cardiovascular disease (81 patients) and a group without a history of cardiovascular disease (27 patients). Results: Patients with cardiovascular antecedents had a higher mortality rate than those without cardiovascular antecedents, presenting severe forms with shorter survival time in the intensive care unit and increased inflammatory evidence. Compared to patients without a history of cardiovascular illness, those with cardiovascular disease had a lower average age, and developed a severe form of COVID-19. Conclusions: Cardiovascular antecedents can worsen the prognosis of patients with COVID-19, requiring a careful screening and multidisciplinary approach.
Collapse
Affiliation(s)
- Luiza Camelia Nechita
- Doctoral School of Biomedical Sciences, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.C.N.); (A.A.S.B.); (R.E.B.)
| | - Mariana Daniela Ignat
- Doctoral School of Biomedical Sciences, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.C.N.); (A.A.S.B.); (R.E.B.)
| | | | - Raisa Eloise Barbu
- Doctoral School of Biomedical Sciences, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.C.N.); (A.A.S.B.); (R.E.B.)
| | - Liliana Baroiu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (D.C.V.); (A.N.); (C.B.)
- ‘Sf. Cuv. Parascheva’ Clinical Hospital of Infectious Diseases, 800179 Galati, Romania;
| | - Doina Carina Voinescu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (D.C.V.); (A.N.); (C.B.)
- ‘Sf. Apostol Andrei’ Clinical Emergency County Hospital, 800578 Galati, Romania
| | - Aurel Nechita
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (D.C.V.); (A.N.); (C.B.)
- ‘Sf. Ioan’ Clinical Hospital for Children, 800487 Galati, Romania;
| | - Mihaela Debita
- ‘Sf. Cuv. Parascheva’ Clinical Hospital of Infectious Diseases, 800179 Galati, Romania;
- Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania
| | - Camelia Busila
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (D.C.V.); (A.N.); (C.B.)
- ‘Sf. Ioan’ Clinical Hospital for Children, 800487 Galati, Romania;
| | - Ioana Anca Stefanopol
- ‘Sf. Ioan’ Clinical Hospital for Children, 800487 Galati, Romania;
- Clinical Surgical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania
| |
Collapse
|
50
|
Bjorgvinsdottir O, Ferguson SJ, Snorradottir BS, Gudjonsson T, Wuertz-Kozak K. The influence of physical and spatial substrate characteristics on endothelial cells. Mater Today Bio 2024; 26:101060. [PMID: 38711934 PMCID: PMC11070711 DOI: 10.1016/j.mtbio.2024.101060] [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: 12/30/2023] [Revised: 03/10/2024] [Accepted: 04/13/2024] [Indexed: 05/08/2024] Open
Abstract
Cardiovascular diseases are a main cause of death worldwide, leading to a growing demand for medical devices to treat this patient group. Central to the engineering of such devices is a good understanding of the biology and physics of cell-surface interactions. In existing blood-contacting devices, such as vascular grafts, the interaction between blood, cells, and material is one of the main limiting factors for their long-term durability. An improved understanding of the material's chemical- and physical properties as well as its structure all play a role in how endothelial cells interact with the material surface. This review provides an overview of how different surface structures influence endothelial cell responses and what is currently known about the underlying mechanisms that guide this behavior. The structures reviewed include decellularized matrices, electrospun fibers, pillars, pits, and grated surfaces.
Collapse
Affiliation(s)
- Oddny Bjorgvinsdottir
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavik, Iceland
| | - Stephen J. Ferguson
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 37 / 39, 8092, Zurich, Switzerland
| | | | - Thorarinn Gudjonsson
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, 101 Reykjavik, Iceland
| | - Karin Wuertz-Kozak
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 160 Lomb Memorial Drive Bldg. 73, Rochester, NY, 14623, USA
| |
Collapse
|