1
|
Gaia de Sousa F, Mendes ACR, de Carvalho LP, Beier SL. Clinical-Diagnostic and Therapeutic Advances in Feline Hypertrophic Cardiomyopathy. Vet Sci 2025; 12:289. [PMID: 40267000 PMCID: PMC11946439 DOI: 10.3390/vetsci12030289] [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: 02/05/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/25/2025] Open
Abstract
A comprehensive literature review was conducted to summarise existing evidence and the latest therapeutic advancements in feline HCM. The disease phenotype is characterised by the development of concentric hypertrophy, which is variable but often asymmetric, primarily affecting the left ventricle. The HCM is marked by diastolic dysfunction, resulting in a reduced intracavitary internal diameter due to the disordered alignment of cardiomyocytes and excessive collagen deposition, which thickens the myocardial tissue and impairs relaxation. There is strong evidence supporting the role of genetic mutations in sarcomeric genes in the development of HCM. Clinical signs vary but are frequently associated with congestive heart failure (CHF) and ATE. Diagnosis is based on a combination of clinical evaluation, laboratory tests, and imaging studies, although certain aspects of the disease remain insufficiently understood and require further research. Therapeutic management focuses on controlling clinical signs, slowing disease progression, and improving both quality of life and life expectancy. However, ongoing studies are essential to refine diagnostic strategies and explore novel treatment options for better disease management.
Collapse
Affiliation(s)
- Felipe Gaia de Sousa
- Department of Veterinary Clinic and Surgery, Veterinary School, Federal University of Minas Gerais—UFMG, Belo Horizonte 31620-295, Minas Gerais, Brazil;
| | - Ana Cristina Ribeiro Mendes
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, Pontifical Catholic University of Minas Gerais—PUC Minas, Belo Horizonte 30140-002, Minas Gerais, Brazil; (A.C.R.M.); (L.P.d.C.)
| | - Luisa Pimenta de Carvalho
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, Pontifical Catholic University of Minas Gerais—PUC Minas, Belo Horizonte 30140-002, Minas Gerais, Brazil; (A.C.R.M.); (L.P.d.C.)
| | - Suzane Lilian Beier
- Department of Veterinary Clinic and Surgery, Veterinary School, Federal University of Minas Gerais—UFMG, Belo Horizonte 31620-295, Minas Gerais, Brazil;
| |
Collapse
|
2
|
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
|
3
|
Moscucci F, Bucciarelli V, Gallina S, Sciomer S, Mattioli AV, Maffei S, Nodari S, Pedrinelli R, Andreozzi P, Basili S. Obstructive sleep apnea syndrome (OSAS) in women: A forgotten cardiovascular risk factor. Maturitas 2025; 193:108170. [PMID: 39708590 DOI: 10.1016/j.maturitas.2024.108170] [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: 07/05/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024]
Abstract
Sleep-disordered breathing is a highly prevalent disorder with negative impact on healthcare systems worldwide. This condition has detrimental effects on cardiovascular health and quality of life, and is frequently associated with a variety of comorbidities, including cardiovascular disease, heart failure, diabetes and atrial fibrillation. Nevertheless, it remains frequently undiagnosed and undertreated, especially in specific populations. Studies on sleep-disordered breathing have been conducted mainly on male patients, and the prevalence and severity of this disorder in women are underestimated. Recently, some clinical and laboratory evidence has highlighted the epidemiological and pathophysiological differences between men and women with sleep-disordered breathing. In this review, we discuss sex-related mechanisms of sleep-disordered breathing in frequently associated disorders, to improve clinical understanding of this condition and to simplify the practical application of targeted interventions. The aim is to improve prognosis among female patients and guarantee a better quality of life and a reduction in healthcare costs.
Collapse
Affiliation(s)
- Federica Moscucci
- Azienda Ospedaliera Universitaria Policlinico Umberto I, DAI of Internal Medicine and Medical Specialties, Viale del Policlinico n.155, 00185 Rome, Italy; Department of Clinical Medicine, Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Valentina Bucciarelli
- Cardiovascular Sciences Department, Azienda Ospedaliero-Universitaria delle Marche, 60126 Ancona, Italy
| | - Sabina Gallina
- Department of Neuroscience, Imaging and Clinical Sciences, "G. D'Annunzi" University, Chieti, Italy.
| | - Susanna Sciomer
- Department of Clinical and Internal Medicine, Anesthesiology and Cardiovascular Sciences, University of Rome "Sapienza", Rome, Italy.
| | | | - Silvia Maffei
- Cardiovascular and Gynaecological Endocrinology Unit, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy.
| | - Savina Nodari
- Department of Cardiology, University of Brescia and ASST "Spedali Civili" Hospital, 25123 Brescia, Italy.
| | - Roberto Pedrinelli
- Cardiac, Thoracic and Vascular Department, University of Pisa, 56126 Pisa, Italy.
| | - Paola Andreozzi
- Predictive Medicine Gender Specificity and Chronicity Unit, Department of Internal Medicine, Endocrine-Metabolic Sciences and Infectious Diseases, Azienda Ospedaliero Universitaria Policlinico Umberto I, SIMI National Directive, Rome, Italy.
| | - Stefania Basili
- Department of Translational and Precision Medicine, Sapienza-University of Rome, Viale del Policlinico 155, Rome 00161, Italy.
| |
Collapse
|
4
|
Donadini MP, Calcaterra F, Romualdi E, Ciceri R, Cancellara A, Lodigiani C, Bacci M, Della Bella S, Ageno W, Mavilio D. The Link Between Venous and Arterial Thrombosis: Is There a Role for Endothelial Dysfunction? Cells 2025; 14:144. [PMID: 39851572 PMCID: PMC11763525 DOI: 10.3390/cells14020144] [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: 11/23/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Venous thromboembolism (VTE) and arterial thrombosis (AT) are distinct yet closely related pathological processes. While traditionally considered separate entities, accumulating evidence suggests that they share common risk factors, such as inflammation and endothelial dysfunction (ED). This review explores the parallels and differences between venous and arterial thrombosis, with particular attention to the role of unprovoked VTE and its potential links to atherosclerosis and systemic inflammation. A key focus is the role of ED, which is emerging as a critical factor in thrombogenesis across both the venous and arterial systems. We examine the current methods for clinically detecting ED, including the use of biomarkers and advanced imaging techniques. Additionally, we discuss novel research avenues, such as the potential of endothelial colony-forming cells and other innovative methodologies, to further unravel the complex mechanisms of thrombosis. Finally, we propose future clinical scenarios where targeting endothelial health could pave the way for more effective prevention and treatment strategies in thrombosis management.
Collapse
Affiliation(s)
- Marco Paolo Donadini
- Department of Medicine and Surgery, Research Center on Thromboembolic Diseases and Antithrombotic Therapies, University of Insubria, 21100 Varese, Italy;
- Centro Trombosi e TAO, Azienda Socio Sanitaria Territoriale dei Sette Laghi, 21100 Varese, Italy;
| | - Francesca Calcaterra
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Erica Romualdi
- Centro Trombosi e TAO, Azienda Socio Sanitaria Territoriale dei Sette Laghi, 21100 Varese, Italy;
| | - Roberta Ciceri
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Assunta Cancellara
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Corrado Lodigiani
- Center for Thrombosis and Hemorrhagic Diseases, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (C.L.); (M.B.)
| | - Monica Bacci
- Center for Thrombosis and Hemorrhagic Diseases, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (C.L.); (M.B.)
| | - Silvia Della Bella
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Walter Ageno
- Department of Medicine and Surgery, Research Center on Thromboembolic Diseases and Antithrombotic Therapies, University of Insubria, 21100 Varese, Italy;
- Department of Internal Medicine, Ospedale Regionale di Bellinzona e Valli, 6500 Bellinzona, Switzerland
| | - Domenico Mavilio
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| |
Collapse
|
5
|
Vostatek R, Ay C. Biological Aging and Venous Thromboembolism: A Review of Telomeres and Beyond. Biomedicines 2024; 13:15. [PMID: 39857599 PMCID: PMC11759860 DOI: 10.3390/biomedicines13010015] [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: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
Although venous thromboembolism (VTE) is the third most common cardiovascular disease, and the risk of VTE increases sharply with advancing age, approximately 40% of VTE cases are currently classified as unprovoked, highlighting the importance of risk factor research. While chronological aging is associated with the risk of VTE, the association with biological aging remains unclear. Biological aging is highly complex, influenced by several dysregulated cellular and biochemical mechanisms. In the last decade, advancements in omics methodologies provided insights into the molecular complexity of biological aging. Techniques such as high-throughput genomics, epigenomics, transcriptomics, proteomics, and metabolomics analyses identified and quantified numerous epigenetic markers, transcripts, proteins, and metabolites. These methods have also revealed the molecular alterations organisms undergo as they age. Despite the progress, there is still a lack of consensus regarding the methods for assessing and validating these biomarkers, and their application lacks standardization. This review gives an overview of biomarkers of biological aging, including telomere length, and their potential role for VTE. Furthermore, we critically examine the advantages and disadvantages of the proposed methods and discuss possible future directions for investigating biological aging in VTE.
Collapse
Affiliation(s)
| | - Cihan Ay
- Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
6
|
de Melo IG, Tavares V, Savva-Bordalo J, Rei M, Liz-Pimenta J, Pereira D, Medeiros R. Endothelial Dysfunction Markers in Ovarian Cancer: VTE Risk and Tumour Prognostic Outcomes. Life (Basel) 2024; 14:1630. [PMID: 39768338 PMCID: PMC11678387 DOI: 10.3390/life14121630] [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: 11/03/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Ovarian cancer (OC) presents daunting lethality rates worldwide, with frequent late-stage diagnosis and chemoresistance, highlighting the need for improved prognostic approaches. Venous thromboembolism (VTE), a major cancer mortality factor, is partially driven by endothelial dysfunction (ED). ED's pro-inflammatory state fosters tumour progression, suggesting a VTE-independent link between ED and cancer. Given this triad's interplay, ED markers may influence OC behaviour and patients' prognosis. Thus, the impact of ED-related genes and single-nucleotide polymorphisms (SNPs) on OC-related VTE and patient thrombogenesis-independent prognosis was investigated. NOS3 upregulation was linked to lower VTE incidence (χ2, p = 0.013), while SELP upregulation was associated with shorter overall survival (log-rank test, p = 0.048). Dismissing patients with VTE before OC diagnosis, SELP rs6136 T allele carriers presented lower progression-free survival (log-rank test, p = 0.038). Nevertheless, due to the SNP minor allele underrepresentation, further investigation is required. Taken together, ED markers seem to exhibit roles that depend on the clinical context, such as tumour-related thrombogenesis or cancer prognosis. Validation with larger cohorts and more in-depth functional studies are needed for data clarification and potential therapeutic strategies exploitation to tackle cancer progression and thrombosis in OC patients.
Collapse
Affiliation(s)
- Inês Guerra de Melo
- Molecular Oncology and Viral Pathology Group, Research Centre of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal; (I.G.d.M.); (V.T.)
- Faculty of Medicine, University of Porto (FMUP), 4200-072 Porto, Portugal;
| | - Valéria Tavares
- Molecular Oncology and Viral Pathology Group, Research Centre of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal; (I.G.d.M.); (V.T.)
- Faculty of Medicine, University of Porto (FMUP), 4200-072 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Joana Savva-Bordalo
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; (J.S.-B.); (D.P.)
| | - Mariana Rei
- Department of Gynaecology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal;
| | - Joana Liz-Pimenta
- Faculty of Medicine, University of Porto (FMUP), 4200-072 Porto, Portugal;
- Department of Medical Oncology, Centro Hospitalar de Trás-os-Montes e Alto Douro (CHTMAD), 5000-508 Vila Real, Portugal
| | - Deolinda Pereira
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; (J.S.-B.); (D.P.)
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Centre of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal; (I.G.d.M.); (V.T.)
- Faculty of Medicine, University of Porto (FMUP), 4200-072 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
- Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
| |
Collapse
|
7
|
Miranda-Mosqueda ML, Ruiz-Oropeza SY, González-Barrios JA, Jaimez R, Peña-Ortega F, Gómez-Acevedo C. Agmatine: An Emerging Approach for Neuroprotection in Recurrent Ischemic Stroke Events in a Murine Model. Drug Dev Res 2024; 85:e70012. [PMID: 39470139 DOI: 10.1002/ddr.70012] [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: 05/08/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024]
Abstract
This study investigates the effect of agmatine on reducing mortality, neurobehavioral alterations, infarct size, and expression of pro-inflammatory cytokines in mice subjected to bilateral carotid thrombosis. Under pentobarbital anesthesia, the left common carotid artery was exposed to 6% FeCl3. Thirty-two days later, the same procedure was performed on the right common carotid artery. Subsequently, Agmatine (100 mg/kg) was administered 15 min after the second procedure, and in another experimental group, the dose of Agmatine was repeated at 72 h. Administration of agmatine extended survival in ischemic animals up to 72 h for the single-dose group and up to 96 h for the repeated-dose group, without significant increases in neurological deficits or infarct area size. This neurobehavioral effect was also observed in sham animals treated with agmatine. In ischemic animals, agmatine administration improved digging behavior and reduced recovery times, consistently shorter in those animals treated with repeated doses. RT-PCR analyses revealed a positive regulation of the cytokine IL-1β in agmatine-treated animals, which has been associated with recovery stages. The results suggest that the observed effect may be attributed to the multiple interactions of agmatine with ischemic cascade events, highlighting its anti-inflammatory role.
Collapse
Affiliation(s)
- M L Miranda-Mosqueda
- Laboratorio de Farmacología Conductual, Departamento de Farmacología, Facultad de Medicina, UNAM, Juriquilla, México
| | - S Y Ruiz-Oropeza
- Laboratorio de Farmacología Conductual, Departamento de Farmacología, Facultad de Medicina, UNAM, Juriquilla, México
| | - J A González-Barrios
- Laboratorio de Medicina Genómica, Hospital Regional 1° de Octubre, ISSSTE, México City, México
| | - R Jaimez
- Laboratorio de Estrógenos y Hemostasis, Departamento de Farmacología, Facultad de Medicina, UNAM, Juriquilla, México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, México
| | - C Gómez-Acevedo
- Laboratorio de Farmacología Conductual, Departamento de Farmacología, Facultad de Medicina, UNAM, Juriquilla, México
| |
Collapse
|
8
|
Zhang Y, Ge Y, Tao L, Liu M, Jia W, Tian X, Jiang P, Cheng Z, Li J, Liu J. Peroxisome proliferator‑activated receptor γ alleviates human umbilical vein endothelial cell injury in deep vein thrombosis by blocking endoplasmic reticulum stress. Exp Ther Med 2024; 28:385. [PMID: 39161618 PMCID: PMC11332125 DOI: 10.3892/etm.2024.12674] [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: 03/31/2023] [Accepted: 02/15/2024] [Indexed: 08/21/2024] Open
Abstract
The present study aimed to explore the role of peroxisome proliferator-activated receptor γ (PPARγ) in the development of deep vein thrombosis (DVT), as well as to discover the potential regulatory mechanism of PPARγ. Human umbilical vein endothelial cells (HUVECs) were treated with modified glycated human serum albumin (M-HSA) to mimic DVT. PPARγ expression and activity were detected using western blot analysis and the corresponding activity detection kit, respectively. Cell Counting Kit-8 and the terminal deoxynucleotidyl-transferase-mediated dUTP nick end labeling assays were employed to detect cell viability and apoptosis, respectively. The levels of thrombosis-related factors and inflammatory cytokines were detected by ELISA. The levels of oxidative stress-related factors were determined by the corresponding commercial kits. In addition, tunicamycin (TM), the agonist of endoplasmic reticulum stress (ERS), was applied to investigate the potential mechanism. The results indicated that M-HSA caused reduced expression and activity of PPARγ in HUVECs; these effects were reversed by PPARγ overexpression, which significantly inhibited M-HSA-induced cell viability loss, cell apoptosis, inflammation and oxidative stress in HUVECs. In addition, ERS was activated following M-HSA stimulation in HUVECs, but was suppressed by PPARγ overexpression. Furthermore, TM partly abolished the protective role of PPARγ overexpression against cell viability loss, cell apoptosis, inflammation and oxidative stress in M-HSA-induced HUVECs. In summary, PPARγ antagonized M-HSA-induced HUVEC injury by suppressing the activation of ERS, which provides a novel strategy for the treatment of DVT.
Collapse
Affiliation(s)
- Yunxin Zhang
- Department of Vascular Surgery, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Yongshuai Ge
- Research Center for Medical Artificial Intelligence, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, P.R. China
| | - Liyuan Tao
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Mingyuan Liu
- Department of Vascular Surgery, Beijing Friendship Hospital, Beijing 100050, P.R. China
| | - Wei Jia
- Department of Vascular Surgery, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Xuan Tian
- Department of Vascular Surgery, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Peng Jiang
- Department of Vascular Surgery, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Zhiyuan Cheng
- Department of Vascular Surgery, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Jinyong Li
- Department of Vascular Surgery, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| | - Jianlong Liu
- Department of Vascular Surgery, Beijing Jishuitan Hospital, Beijing 100035, P.R. China
| |
Collapse
|
9
|
Panchagnula N, Brasher WP. Hyperglycemia and Venous Thromboembolism. Diagnostics (Basel) 2024; 14:1994. [PMID: 39272778 PMCID: PMC11393887 DOI: 10.3390/diagnostics14171994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Patients with diabetes mellitus (DM) have chronically increased blood glucose and multiple physiologic alterations that place them at elevated risk for vascular disease. Traditionally, this vascular risk has mainly referred to chronic atherosclerosis and embolic arterial disease. Retrospective studies have suggested an increased risk of a pulmonary embolism (PE) and deep vein thrombosis (DVT), collectively termed venous thromboembolism (VTE), in patients with DM, but this association has been difficult to demonstrate with comorbidities such as obesity in meta-analysis. Clinical studies have demonstrated worse outcomes for patients with DM who suffer from VTE. In vitro studies show multiple physiologic abnormalities with chronic inflammation, endothelial dysfunction, dysfunction in the coagulation cascade, as well as other changes that drive a vicious cycle of hypercoagulability. Aggressive medical management of DM can improve vascular outcomes, and some anti-hyperglycemic therapies may modify VTE risk as well. Anticoagulation strategies are similar for patients with DM, but with some added considerations, such as high rates of comorbid renal dysfunction. More research is needed to definitively categorize DM as a risk factor for VTE and elucidate specific therapeutic strategies.
Collapse
Affiliation(s)
- Neha Panchagnula
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - William Philip Brasher
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
10
|
de Melo IG, Tavares V, Pereira D, Medeiros R. Contribution of Endothelial Dysfunction to Cancer Susceptibility and Progression: A Comprehensive Narrative Review on the Genetic Risk Component. Curr Issues Mol Biol 2024; 46:4845-4873. [PMID: 38785560 PMCID: PMC11120512 DOI: 10.3390/cimb46050292] [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: 04/14/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Venous thromboembolism (VTE) is a challenging clinical obstacle in oncological settings, marked by elevated incidence rates and resulting morbidity and mortality. In the context of cancer-associated thrombosis (CAT), endothelial dysfunction (ED) plays a crucial role in promoting a pro-thrombotic environment as endothelial cells lose their ability to regulate blood flow and coagulation. Moreover, emerging research suggests that this disorder may not only contribute to CAT but also impact tumorigenesis itself. Indeed, a dysfunctional endothelium may promote resistance to therapy and favour tumour progression and dissemination. While extensive research has elucidated the multifaceted mechanisms of ED pathogenesis, the genetic component remains a focal point of investigation. This comprehensive narrative review thus delves into the genetic landscape of ED and its potential ramifications on cancer progression. A thorough examination of genetic variants, specifically polymorphisms, within key genes involved in ED pathogenesis, namely eNOS, EDN1, ACE, AGT, F2, SELP, SELE, VWF, ICAM1, and VCAM1, was conducted. Overall, these polymorphisms seem to play a context-dependent role, exerting both oncogenic and tumour suppressor effects depending on the tumour and other environmental factors. In-depth studies are needed to uncover the mechanisms connecting these DNA variations to the pathogenesis of malignant diseases.
Collapse
Affiliation(s)
- Inês Guerra de Melo
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Dep., Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal; (I.G.d.M.); (V.T.)
- Faculty of Medicine of University of Porto (FMUP), 4200-072 Porto, Portugal
| | - Valéria Tavares
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Dep., Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal; (I.G.d.M.); (V.T.)
- Faculty of Medicine of University of Porto (FMUP), 4200-072 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Deolinda Pereira
- Oncology Department, Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal;
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Dep., Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal; (I.G.d.M.); (V.T.)
- Faculty of Medicine of University of Porto (FMUP), 4200-072 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
- Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
| |
Collapse
|
11
|
Krarup KB, Krarup HB, Mørk M, Lundbye-Christensen S, Handberg A, Nguyen HTT, Pedersen IS, Kristensen SR. Are Gamers Prone to eThrombosis during Long Gaming Sessions? Life (Basel) 2024; 14:525. [PMID: 38672795 PMCID: PMC11051545 DOI: 10.3390/life14040525] [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: 03/21/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
During the last two decades, several cases of venous thrombosis (VTE) after a prolonged period at a computer have been described, denominated as "eThrombosis". Video gaming on a computer has become very popular and can be a social activity where several players gather to play against each other or in a virtual environment for several days ("LAN (i.e., Local Area Network) parties") where the participants are sedentary and consuming calorie-rich food items. The aim of this study was to investigate potential coagulation activation during a 42 h LAN party. Nine male gamers volunteered for the LAN party. Citrated blood was sampled before and every 6 h, and plasma was analyzed for thrombin generation, thrombin-antithrombin complexes (TAT), prothrombin fragment 1 + 2 (F1 + 2), and D-dimer. Thrombin generation increased slightly but not significantly during the LAN party, whereas the coagulation activation markers were unchanged. These results do not indicate that the coagulation system is activated significantly during 42 h of gaming with minimal physical activity. Although increased activity cannot be excluded, it does not directly indicate a risk of VTE in general.
Collapse
Affiliation(s)
- Kasper B. Krarup
- Department of Geriatrics, Aalborg University Hospital, 9000 Aalborg, Denmark;
- Sports Medicine Center, Region Hospital North Jutland, 9900 Frederikshavn, Denmark
| | - Henrik B. Krarup
- Department of Molecular Diagnostics, Aalborg University Hospital, 9000 Aalborg, Denmark; (H.B.K.); (H.T.T.N.); (I.S.P.)
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark; (S.L.-C.)
| | - Morten Mørk
- Department of Clinical Biochemistry, Aalborg University Hospital, 9000 Aalborg, Denmark;
| | | | - Aase Handberg
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark; (S.L.-C.)
- Department of Clinical Biochemistry, Aalborg University Hospital, 9000 Aalborg, Denmark;
| | - Hien T. T. Nguyen
- Department of Molecular Diagnostics, Aalborg University Hospital, 9000 Aalborg, Denmark; (H.B.K.); (H.T.T.N.); (I.S.P.)
| | - Inge S. Pedersen
- Department of Molecular Diagnostics, Aalborg University Hospital, 9000 Aalborg, Denmark; (H.B.K.); (H.T.T.N.); (I.S.P.)
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark; (S.L.-C.)
| | - Søren R. Kristensen
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark; (S.L.-C.)
- Department of Clinical Biochemistry, Aalborg University Hospital, 9000 Aalborg, Denmark;
| |
Collapse
|
12
|
Liu F, Zhai Q. Expression level of neutrophil extracellular traps in peripheral blood of patients with chronic heart failure complicated with venous thrombosis and its clinical significance. J Cardiothorac Surg 2024; 19:129. [PMID: 38491551 PMCID: PMC10941499 DOI: 10.1186/s13019-024-02506-3] [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: 03/16/2023] [Accepted: 01/18/2024] [Indexed: 03/18/2024] Open
Abstract
OBJECTIVE Previous studies have reported that neutrophil extracellular traps (NETs) have been identified to be involved in thrombosis, but the clinical value in chronic heart failure (CHF) patients with venous thrombosis is unclear. This study focused on the expression level of NETs in the peripheral blood of patients with CHF complicated with venous thrombosis and its clinical value. METHODS 80 patients with CHF were included and divided into 2 groups according to the occurrence of venous thrombosis, and the expression levels of NETs in peripheral venous blood and lesion veins of the patients were detected through fluorescent staining. Myeloperoxidase-DNA (MPO-DNA) and citrullinated histone H3 (CitH3), markers of NETs, were detected by enzyme linked immunosorbent assay kit. The receiver operating characteristic (ROC) curve was used to analyze the value of peripheral venous blood NETs in the diagnosis of venous thrombosis in CHF patients, while the relationship between NETs in peripheral and lesion veins was analyzed by a unitary linear regression model. RESULTS The results showed that the concentration of NETs, MPO-DNA, and CitH3 in CHF patients combined with venous thrombosis was markedly higher than that in patients without venous thrombosis, and the concentration of NETs, MPO-DNA, and CitH3 in lesion venous blood was notably higher than that in peripheral venous blood. Binary logistics regression analysis showed that NETs in peripheral venous blood were an independent risk factor for venous thrombosis in patients with heart failure. The unitary linear regression model fitted well, indicating a notable positive correlation between NETs concentrations in peripheral and lesion veins. The area under the ROC curve for diagnosing venous thrombosis was 0.85, indicating that peripheral blood NETs concentration levels could effectively predict venous thrombosis in CHF patients. CONCLUSION The expression level of NETs was high in the peripheral blood of CHF patients combined with venous thrombosis and was the highest in lesion venous blood. NETs levels in peripheral blood had the value of diagnosing venous thrombosis in CHF patients, and the concentrations of NETs in peripheral and lesion veins are markedly positively correlated.
Collapse
Affiliation(s)
- Fang Liu
- Medical Lab, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, 710038, Shaanxi, China
| | - Qian Zhai
- Department of Blood test, Xi'an Blood Center, Shaanxi Blood Center, No.407 Zhuque Street, Yanta District, Xi'an, 710061, Shaanxi Province, China.
| |
Collapse
|
13
|
Tsai HR, Lin YJ, Yeh JI, Huang YC, Liu PPS, Peng CCH, Hsu JY, Lee YC, Loh CH, Lin SM, Huang HK. Sodium-glucose co-transporter-2 inhibitors and the risk of venous thromboembolism: A nationwide population-based study and meta-analysis. Diabetes Metab Res Rev 2024; 40:e3739. [PMID: 37862117 DOI: 10.1002/dmrr.3739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/04/2023] [Accepted: 09/08/2023] [Indexed: 10/22/2023]
Abstract
AIMS Sodium-glucose cotransporter-2 inhibitors (SGLT-2i) have off-target effects on haemoconcentration and anti-inflammation. The impact of SGLT-2i on the risk of venous thromboembolism (VTE) in patients with diabetes mellitus (DM) remains unclear. This study aimed to evaluate the risk of newly diagnosed VTE in patients with DM using SGLT-2i in comparison to dipeptidyl peptidase-4 inhibitors (DPP-4i) or glucagon-like peptide-1 receptor agonists (GLP-1RA). MATERIALS AND METHODS In this nationwide retrospective cohort study, we used data from Taiwan's National Health Insurance Research Database. Patients with diabetes aged 20 years or older who received SGLT-2i, DPP-4i, or GLP-1RA between 1 May 2016, and 31 December 2020, were included. The risks of VTE in SGLT-2i users were compared with those of DPP-4i and GLP-1RA users. A Cox regression model with stabilised inverse probability of treatment weighting was used to calculate hazard ratio (HR) for VTE risk. Additionally, a meta-analysis of relevant articles published before 23 May 2023, was conducted. RESULTS Data from 136,530 SGLT-2i, 598,280 DPP-4i, and 5760 GLP-1RA users were analysed. SGLT-2i use was associated with a lower risk of VTE than DPP-4i (HR, 0.70; 95% CI, 0.59-0.84; p < 0·001), but not with GLP-1RA (HR, 1.39; 95% CI, 0.32-5.94; p = 0.66). Our meta-analysis further supported these findings (SGLT-2i vs. DPP-4i: HR, 0.71; 95% CI, 0.62-0.82; p < 0·001; SGLT-2i vs. GLP-1RA: HR, 0.91; 95% CI, 0.73-1.15; p = 0.43), suggesting the robustness of our retrospective analysis. CONCLUSIONS In patients with DM, SGLT-2i was associated with a lower risk of VTE compared to DPP-4i, but not GLP-1RA.
Collapse
Affiliation(s)
- Hou-Ren Tsai
- Department of Ophthalmology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Yu-Jie Lin
- Health Information Center, Tzu Chi University, Hualien, Taiwan
| | - Jih-I Yeh
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Family Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Yu-Chi Huang
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Peter Pin-Sung Liu
- Center for Aging and Health, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Carol Chiung-Hui Peng
- Section of Endocrinology, Diabetes, Nutrition & Weight Management, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Jin-Yi Hsu
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Center for Aging and Health, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Yuan-Chieh Lee
- Department of Ophthalmology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Ophthalmology and Visual Science, Tzu Chi University, Hualien, Taiwan
| | - Ching-Hui Loh
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Center for Aging and Health, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Shu-Man Lin
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Physical Medicine and Rehabilitation, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Huei-Kai Huang
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Family Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
14
|
Badulescu OV, Badescu MC, Bojan IB, Vladeanu M, Filip N, Dobreanu S, Tudor R, Ciuntu BM, Tanevski A, Ciocoiu M. Thrombotic Disease in Hemophilic Patients: Is This a Paradox in a State of Hypocoagulability? Diagnostics (Basel) 2024; 14:286. [PMID: 38337802 PMCID: PMC10854955 DOI: 10.3390/diagnostics14030286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Hemophilia patients have a deficiency in or dysfunction of clotting factors, which can lead to a bleeding tendency. However, paradoxically, some hemophilia patients may also be at an increased risk of developing thrombotic events such as deep vein thrombosis or pulmonary embolism. The pathophysiology of thrombosis in hemophilia patients is not fully understood, but it is thought to involve a complex interplay of various factors, including the severity of the hemophilia, the presence of other risk factors such as obesity, smoking, or the use of hormonal therapies, and the presence of certain genetic mutations that increase the risk of thrombosis. In addition, it has been suggested that the use of clotting factor replacement therapy, which is a standard treatment for hemophilia, may also contribute to the development of thrombosis in some cases.
Collapse
Affiliation(s)
- Oana Viola Badulescu
- Department of Pathophysiology, Morpho-Functional Sciences (II), Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (O.V.B.); (M.V.); (M.C.)
| | - Minerva Codruta Badescu
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Iris Bararu Bojan
- Department of Pathophysiology, Morpho-Functional Sciences (II), Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (O.V.B.); (M.V.); (M.C.)
| | - Maria Vladeanu
- Department of Pathophysiology, Morpho-Functional Sciences (II), Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (O.V.B.); (M.V.); (M.C.)
| | - Nina Filip
- Department of Biochemistry, Morpho-Functional Sciences (II), Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Stefan Dobreanu
- Institute of Cardiovascular Diseases, G.I.M. Georgescu, 700503 Iasi, Romania
| | - Razvan Tudor
- Department of Orthopedics and Traumatology, Surgical Science (II), Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Bogdan-Mihnea Ciuntu
- Department of General Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (B.-M.C.); (A.T.)
| | - Adelina Tanevski
- Department of General Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (B.-M.C.); (A.T.)
| | - Manuela Ciocoiu
- Department of Pathophysiology, Morpho-Functional Sciences (II), Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (O.V.B.); (M.V.); (M.C.)
| |
Collapse
|
15
|
Wang T, Yong Y, Ge X, Wang J. A computational model-based study on the feasibility of predicting post-splenectomy thrombosis using hemodynamic metrics. Front Bioeng Biotechnol 2024; 11:1276999. [PMID: 38274008 PMCID: PMC10808826 DOI: 10.3389/fbioe.2023.1276999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/30/2023] [Indexed: 01/27/2024] Open
Abstract
For portal hypertensive patients with splenomegaly and hypersplenism, splenectomy is an effective surgery to relieve the complications. However, patients who have undergone splenectomy often suffer from portal venous system thrombosis, a sequela that requires prophylaxis and timely treatment to avoid deterioration and death. The aim of this study is to investigate the feasibility of predicting post-splenectomy thrombosis using hemodynamic metrics based on computational models. First, 15 portal hypertensive patients who had undergone splenectomy were enrolled, and their preoperative clinical data and postoperative follow-up results were collected. Next, computational models of the portal venous system were constructed based on the preoperative computed tomography angiography images and ultrasound-measured flow velocities. On this basis, splenectomy was mimicked and the postoperative area of low wall shear stress (ALWSS) was simulated for each patient-specific model. Finally, model-simulated ALWSS was statistically compared with the patient follow-up results to investigate the feasibility of predicting post-splenectomy thrombosis using hemodynamic metrics. Results showed that ALWSS could predict the occurrence of post-splenectomy thrombosis with the area under the receiver operating characteristic curve (AUC) equal to 0.75. Moreover, statistical analysis implied that the diameter of the splenic vein is positively correlated with ALWSS (r = 0.883, p < 0.0001), and the anatomical structures of the portal venous system also influence the ALWSS. These findings demonstrated that the computational model-based hemodynamic metric ALWSS, which is associated with the anatomorphological features of the portal venous system, is capable of predicting the occurrence of post-splenectomy thrombosis, promoting better prophylaxis and postoperative management for portal hypertensive patients receiving splenectomy.
Collapse
Affiliation(s)
- Tianqi Wang
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China
- School of Mechanical Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yan Yong
- College of Science, University of Shanghai for Science and Technology, Shanghai, China
| | - Xinyang Ge
- College of Mathematical Medicine, Zhejiang Normal University, Jinhua, China
| | - Jitao Wang
- Department of Hepatobiliary Surgery, Xingtai Institute of Cancer Control, Xingtai, China
| |
Collapse
|
16
|
Rockholt MM, Naddi L, Badri AM, Englund E, Kander T. Macro- and microscopic changes in veins with short-term central venous catheters: an observational autopsy study. BMC Anesthesiol 2024; 24:5. [PMID: 38166620 PMCID: PMC10759750 DOI: 10.1186/s12871-023-02380-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Centrally inserted central catheters (CICCs) are indispensable in modern healthcare, but unfortunately, come with complications. Catheter-related thrombosis is a well-known complication reported to occur in 5-30% of patients with CICC. There is a paucity of studies that report the incidence of catheter-related thrombosis after the introduction of real-time ultrasound insertion guidance as clinical practice. This study aimed to demonstrate any pathological macro- or microscopic changes in the vein wall associated with CICCs. METHODS The study was approved by the Swedish Ethical Review Authority and was conducted at a large university hospital. The study included 12 patients with a short-term CICC who were subject to autopsies. Vessels with inserted catheters were macroscopically and microscopically examined. RESULTS In total, seven female and five male patients with a median age of 70 (interquartile range 63-76) were included. With one exception, all patients received routine thromboprophylaxis throughout the period with CICC. Most inserted CICCs were 9.5 French (54%) and were inserted in the internal jugular vein (92%). The median time with CICC was seven days (interquartile range 1.8-20). At autopsy, thrombi were observed in all cases (100%), macroscopically and microscopically, attached to the distal portion of the CICC and/or the adjacent vessel wall. Inflammatory changes in the vessel walls were seen in all cases, and varying degrees of fibrosis were demonstrated in eight cases (67%). CONCLUSIONS This autopsy study demonstrated that catheter-related thrombus formation with adjacent inflammatory and fibrotic vessel wall thickening was very common, despite a limited period of catheter use. The consequences of these findings are important, as thrombi may cause pulmonary embolism and possibly lead to catheter-related infections, and since inflammatory and fibrotic vessel wall thickening may evolve into chronic venous stenosis. Furthermore, the findings are a cause of concern, as CICCs are indispensable in modern healthcare and complications may be masked by the general disease that was the indication for CICC insertion.
Collapse
Affiliation(s)
- Mika M Rockholt
- Department of Intensive and Perioperative Care, Skåne University Hospital, 221 85, Lund, Sweden
- Department of Anesthesiology, Perioperative Care and Pain Medicine, NYU Langone Health, NYC, NY, USA
- Department of Clinical Sciences, Lund University, Box 117, 221 00, Lund, Sweden
| | - Leila Naddi
- Department of Intensive and Perioperative Care, Skåne University Hospital, 221 85, Lund, Sweden
- Department of Clinical Sciences, Lund University, Box 117, 221 00, Lund, Sweden
| | - Ahmed M Badri
- Department of Intensive and Perioperative Care, Skåne University Hospital, 221 85, Lund, Sweden
- Department of Anaesthesiology and Critical Care, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Elisabet Englund
- Department of Clinical Sciences, Lund University, Box 117, 221 00, Lund, Sweden
- Department of Genetics, Pathology and Molecular Diagnostics, Region Skåne, Sweden
| | - Thomas Kander
- Department of Intensive and Perioperative Care, Skåne University Hospital, 221 85, Lund, Sweden.
- Department of Clinical Sciences, Lund University, Box 117, 221 00, Lund, Sweden.
| |
Collapse
|
17
|
Poredoš P, Mukherjee D, Blinc A. Statins and Venous Thromboembolic Disease - Where are we Now? Curr Vasc Pharmacol 2024; 22:297-300. [PMID: 38441024 DOI: 10.2174/0115701611308323240229050237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 01/01/1970] [Accepted: 02/12/2024] [Indexed: 03/06/2024]
Abstract
Classical risk factors for atherosclerosis also play a role in the pathogenesis of venous thromboembolism (VTE). Low-density lipoprotein cholesterol has prothrombotic and endothelium- deteriorating effects which are not limited to the arterial system. The association between hypercholesterolemia and VTE has been established, but the benefits of statins in the prevention of VTE assessed by observation studies seemed equivocal. The large, randomized trial Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin (JUPITER) recorded the occurrence of VTE as a protocol-specified endpoint and reported a reduced incidence of VTE among subjects taking 20 mg of rosuvastatin daily vs placebo (hazard ratio 0.57; 95% confidence interval 0.37-0.86; p=0.007). Similar results were confirmed by meta-analyses of observation studies and randomized trials. Recently, a Mendelian randomization study that took the presence of gene variants coding for less efficient hydroxymethyl-glutaryl coenzyme A reductase activity as a proxy for statin treatment, confirmed a small, but significant negative association between the score of selected genetic polymorphisms and the incidence of VTE. However, since the protective effects of statins are limited, they should not be substituted for guideline-recommended VTE prophylaxis or anticoagulation treatment.
Collapse
Affiliation(s)
- Pavel Poredoš
- Department of Vascular Diseases, University Medical Centre Ljubljana, Slovenia
| | - Debabrata Mukherjee
- Department of Internal Medicine, Texas Tech University Health Sciences Center at El Paso, Texas 79905, USA
| | - Aleš Blinc
- Department of Vascular Diseases, University Medical Centre Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Slovenia
| |
Collapse
|
18
|
Marques IS, Tavares V, Savva-Bordalo J, Rei M, Liz-Pimenta J, de Melo IG, Assis J, Pereira D, Medeiros R. Long Non-Coding RNAs: Bridging Cancer-Associated Thrombosis and Clinical Outcome of Ovarian Cancer Patients. Int J Mol Sci 2023; 25:140. [PMID: 38203310 PMCID: PMC10778953 DOI: 10.3390/ijms25010140] [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: 11/01/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Ovarian cancer (OC) and venous thromboembolism (VTE) have a close relationship, in which tumour cells surpass the haemostatic system to drive cancer progression. Long non-coding RNAs (lncRNAs) have been implicated in VTE pathogenesis, yet their roles in cancer-associated thrombosis (CAT) and their prognostic value are unexplored. Understanding how these lncRNAs influence venous thrombogenesis and ovarian tumorigenesis may lead to the identification of valuable biomarkers for VTE and OC management. Thus, this study evaluated the impact of five lncRNAs, namely MALAT1, TUG1, NEAT1, XIST and MEG8, on a cohort of 40 OC patients. Patients who developed VTE after OC diagnosis had worse overall survival compared to their counterparts (log-rank test, p = 0.028). Elevated pre-chemotherapy MEG8 levels in peripheral blood cells (PBCs) predicted VTE after OC diagnosis (Mann-Whitney U test, p = 0.037; Χ2 test, p = 0.033). In opposition, its low levels were linked to a higher risk of OC progression (adjusted hazard ratio (aHR) = 3.00; p = 0.039). Furthermore, low pre-chemotherapy NEAT1 levels in PBCs were associated with a higher risk of death (aHR = 6.25; p = 0.008). As for the remaining lncRNAs, no significant association with VTE incidence, OC progression or related mortality was observed. Future investigation with external validation in larger cohorts is needed to dissect the implications of the evaluated lncRNAs in OC patients.
Collapse
Affiliation(s)
- Inês Soares Marques
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; (I.S.M.); (V.T.); (I.G.d.M.)
- Faculty of Sciences of the University of Porto (FCUP), 4169-007 Porto, Portugal
| | - Valéria Tavares
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; (I.S.M.); (V.T.); (I.G.d.M.)
- Faculty of Medicine of the University of Porto (FMUP), 4200-072 Porto, Portugal;
- Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Joana Savva-Bordalo
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; (J.S.-B.); (D.P.)
| | - Mariana Rei
- Department of Gynaecology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal;
| | - Joana Liz-Pimenta
- Faculty of Medicine of the University of Porto (FMUP), 4200-072 Porto, Portugal;
- Department of Medical Oncology, Centro Hospitalar de Trás-os-Montes e Alto Douro (CHTMAD), 5000-508 Vila Real, Portugal
| | - Inês Guerra de Melo
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; (I.S.M.); (V.T.); (I.G.d.M.)
- Faculty of Medicine of the University of Porto (FMUP), 4200-072 Porto, Portugal;
| | - Joana Assis
- Clinical Research Unit, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal;
| | - Deolinda Pereira
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; (J.S.-B.); (D.P.)
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; (I.S.M.); (V.T.); (I.G.d.M.)
- Faculty of Medicine of the University of Porto (FMUP), 4200-072 Porto, Portugal;
- Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
- Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
| |
Collapse
|
19
|
Tang P, Wang Y, Yang X, Wu Z, Chen W, Ye Y, Jiang Y, Lin L, Lin B, Lin B. Protective Role of Endothelial SIRT1 in Deep Vein Thrombosis and Hypoxia-induced Endothelial Dysfunction Mediated by NF-κB Deacetylation. Inflammation 2023; 46:1887-1900. [PMID: 37354359 DOI: 10.1007/s10753-023-01848-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/26/2023]
Abstract
Venous hypoxia is considered as the major pathogenetic mechanism linking blood flow stagnancy with deep vein thrombosis (DVT). Our previous study showed that activating SIRT1 may attenuate inferior vena cava (IVC) stenosis-induced DVT in rats. This study was aimed to investigate the role of endothelial SIRT1 in DVT and hypoxia-induced endothelial dysfunction as well as the underlying mechanism. Protein profiling of IVCs and blood plasma of DVT rats induced by IVC stenosis was analysed by 4D Label free proteomics analysis. To verify the independent role of SIRT1 in DVT and oxygen-glucose deprivation (OGD)-induced endothelial dysfunction, SIRT1 specific activator SRT1720 and SIRT1 knockdown in both local IVCs and endothelial cells were employed. Moreover, the role of the NF-κB were investigated using NF-κB inhibitor caffeic acid phenethyl ester (CAPE). SRT1720 significantly inhibited thrombus burden, leukocytes infiltration, protein expressions of cell adhesion molecules and chemokines, as well as acetylation level of NF-κB/p65 in wild DVT rats, while these protective effects of SRT1720 were abolished in rats with SIRT1 knockdown in local IVCs. In vitro, SRT1720 protected endothelial cells against OGD-induced dysfunction characterized with enhanced adhesion of monocytes as well as the protein expressions of cell adhesion molecules and chemokines, whereas these protective effects of SRT1720 were vanished by SIRT1 stable knockdown. Furthermore, CAPE attenuated endothelial cell dysfunction and abolished these effects of SIRT1 knockdown. Collectively, these data suggested that endothelial SIRT1 plays an independent role in ameliorating hypoxia-induced endothelial dysfunction and thrombotic inflammation in DVT, and this effect is mediated by NF-κB deacetylation.
Collapse
Affiliation(s)
- Ping Tang
- Experimental Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yiting Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xinrong Yang
- Experimental Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhongrui Wu
- Experimental Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Wenpei Chen
- Experimental Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yuxin Ye
- Experimental Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yong Jiang
- Experimental Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Liuqing Lin
- Experimental Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Bingqing Lin
- College of Mathematics and Statistics, Shenzhen University, Shenzhen, 518060, China.
| | - Baoqin Lin
- Experimental Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
20
|
Maqbool KU, Arsh H, Kumar D, Veena F, Punshi AK, Payal F, Kumar S, Kumar S, Rani D, Malik J. Cardiovascular Manifestations of Human Monkeypox Virus: An Updated Review. Curr Probl Cardiol 2023; 48:101869. [PMID: 37302648 PMCID: PMC10251728 DOI: 10.1016/j.cpcardiol.2023.101869] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/13/2023]
Abstract
Cardiovascular manifestations in human monkeypox virus (MPXV) infection has gained increasing recognition as significant complications with both social and clinical implications. Myocarditis, viral pericarditis, heart failure, and arrhythmias can occur, leading to adverse effects on individuals' health and quality of life. Understanding the detailed pathophysiology of these cardiovascular manifestations is essential for improved diagnosis and management. The social implications of these cardiovascular complications are multifaceted, ranging from public health concerns and the impact on individuals' quality of life to psychological distress and social stigma. Clinically, diagnosing, and managing these complications present challenges, requiring a multidisciplinary approach and specialized care. The burden on healthcare resources necessitates preparedness and resource allocation to effectively address these complications. We delve into the pathophysiological mechanisms involved, including viral-induced cardiac damage, immune response, and inflammatory processes. Additionally, we explore the types of cardiovascular manifestations and their clinical presentations. Addressing cardiovascular manifestations' social and clinical implications in MPXV infection requires a comprehensive approach involving healthcare professionals, public health authorities, and communities. By prioritizing research, enhancing diagnosis and treatment strategies, and promoting preventive measures, we can mitigate the impact of these complications, improve patient care, and protect public health.
Collapse
Affiliation(s)
| | - Hina Arsh
- Department of Medicine, THQ Hospital, Pasrur, Pakistan
| | - Deepak Kumar
- Department of Medicine, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Fnu Veena
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro, Pakistan
| | | | - Fnu Payal
- Department of Medicine, Shaheed Mohtarma Benazir Bhutto University, Larkana, Pakistan
| | - Sameet Kumar
- Department of Medicine, Shaheed Mohtarma Benazir Bhutto University, Larkana, Pakistan
| | - Saroop Kumar
- Department of Medicine, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Deepa Rani
- Department of Medicine, Shaheed Mohtarma Benazir Bhutto University, Larkana, Pakistan
| | - Jahanzeb Malik
- Department of Cardiovascular Research, Cardiovascular Analytics Group, Islamabad, Pakistan.
| |
Collapse
|
21
|
Natae SF, Merzah MA, Sándor J, Ádány R, Bereczky Z, Fiatal S. A combination of strongly associated prothrombotic single nucleotide polymorphisms could efficiently predict venous thrombosis risk. Front Cardiovasc Med 2023; 10:1224462. [PMID: 37745125 PMCID: PMC10511882 DOI: 10.3389/fcvm.2023.1224462] [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: 05/17/2023] [Accepted: 08/03/2023] [Indexed: 09/26/2023] Open
Abstract
Background Venous thrombosis (VT) is multifactorial trait that contributes to the global burden of cardiovascular diseases. Although abundant single nucleotide polymorphisms (SNPs) provoke the susceptibility of an individual to VT, research has found that the five most strongly associated SNPs, namely, rs6025 (F5 Leiden), rs2066865 (FGG), rs2036914 (F11), rs8176719 (ABO), and rs1799963 (F2), play the greatest role. Association and risk prediction models are rarely established by using merely the five strongly associated SNPs. This study aims to explore the combined VT risk predictability of the five SNPs and well-known non-genetic VT risk factors such as aging and obesity in the Hungarian population. Methods SNPs were genotyped in the VT group (n = 298) and control group (n = 400). Associations were established using standard genetic models. Genetic risk scores (GRS) [unweighted GRS (unGRS), weighted GRS (wGRS)] were also computed. Correspondingly, the areas under the receiver operating characteristic curves (AUCs) for genetic and non-genetic risk factors were estimated to explore their VT risk predictability in the study population. Results rs6025 was the most prevalent VT risk allele in the Hungarian population. Its risk allele frequency was 3.52-fold higher in the VT group than that in the control group [adjusted odds ratio (AOR) = 3.52, 95% CI: 2.50-4.95]. Using all genetic models, we found that rs6025 and rs2036914 remained significantly associated with VT risk after multiple correction testing was performed. However, rs8176719 remained statistically significant only in the multiplicative (AOR = 1.33, 95% CI: 1.07-1.64) and genotypic models (AOR = 1.77, 95% CI: 1.14-2.73). In addition, rs2066865 lost its significant association with VT risk after multiple correction testing was performed. Conversely, the prothrombin mutation (rs1799963) did not show any significant association. The AUC of Leiden mutation (rs6025) showed better discriminative accuracy than that of other SNPs (AUC = 0.62, 95% CI: 0.57-0.66). The wGRS was a better predictor for VT than the unGRS (AUC = 0.67 vs. 0.65). Furthermore, combining genetic and non-genetic VT risk factors significantly increased the AUC to 0.89 with statistically significant differences (Z = 3.924, p < 0.0001). Conclusions Our study revealed that the five strongly associated SNPs combined with non-genetic factors could efficiently predict individual VT risk susceptibility. The combined model was the best predictor of VT risk, so stratifying high-risk individuals based on their genetic profiling and well-known non-modifiable VT risk factors was important for the effective and efficient utilization of VT risk preventive and control measures. Furthermore, we urged further study that compares the VT risk predictability in the Hungarian population using the formerly discovered VT SNPs with the novel strongly associated VT SNPs.
Collapse
Affiliation(s)
- Shewaye Fituma Natae
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Health Sciences, University of Debrecen, Debrecen, Hungary
| | - Mohammed Abdulridha Merzah
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Health Sciences, University of Debrecen, Debrecen, Hungary
| | - János Sándor
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Public Health Research Group, Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Róza Ádány
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsuzsanna Bereczky
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilvia Fiatal
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
22
|
Giuli L, Pallozzi M, Venturini G, Gasbarrini A, Ponziani FR, Santopaolo F. Molecular Mechanisms Underlying Vascular Liver Diseases: Focus on Thrombosis. Int J Mol Sci 2023; 24:12754. [PMID: 37628933 PMCID: PMC10454315 DOI: 10.3390/ijms241612754] [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: 07/15/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Vascular liver disorders (VLDs) comprise a wide spectrum of clinical-pathological entities that primarily affect the hepatic vascular system of both cirrhotic and non-cirrhotic patients. VLDs more frequently involve the portal and the hepatic veins, as well as liver sinusoids, resulting in an imbalance of liver homeostasis with serious consequences, such as the development of portal hypertension and liver fibrosis. Surprisingly, many VLDs are characterized by a prothrombotic phenotype. The molecular mechanisms that cause thrombosis in VLD are only partially explained by the alteration in the Virchow's triad (hypercoagulability, blood stasis, and endothelial damage) and nowadays their pathogenesis is incompletely described and understood. Studies about this topic have been hampered by the low incidence of VLDs in the general population and by the absence of suitable animal models. Recently, the role of coagulation imbalance in liver disease has been postulated as one of the main mechanisms linked to fibrogenesis, so a novel interest in vascular alterations of the liver has been renewed. This review provides a detailed analysis of the current knowledge of molecular mechanisms of VLD. We also focus on the promising role of anticoagulation as a strategy to prevent liver complications and to improve the outcome of these patients.
Collapse
Affiliation(s)
- Lucia Giuli
- Hepatology Unit, CEMAD Centro Malattie Dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (M.P.); (G.V.); (F.R.P.); (F.S.)
| | - Maria Pallozzi
- Hepatology Unit, CEMAD Centro Malattie Dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (M.P.); (G.V.); (F.R.P.); (F.S.)
| | - Giulia Venturini
- Hepatology Unit, CEMAD Centro Malattie Dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (M.P.); (G.V.); (F.R.P.); (F.S.)
| | - Antonio Gasbarrini
- Hepatology Unit, CEMAD Centro Malattie Dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (M.P.); (G.V.); (F.R.P.); (F.S.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Hepatology Unit, CEMAD Centro Malattie Dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (M.P.); (G.V.); (F.R.P.); (F.S.)
| | - Francesco Santopaolo
- Hepatology Unit, CEMAD Centro Malattie Dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (M.P.); (G.V.); (F.R.P.); (F.S.)
| |
Collapse
|
23
|
Marques IS, Tavares V, Neto BV, Mota INR, Pereira D, Medeiros R. Long Non-Coding RNAs in Venous Thromboembolism: Where Do We Stand? Int J Mol Sci 2023; 24:12103. [PMID: 37569483 PMCID: PMC10418965 DOI: 10.3390/ijms241512103] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Venous thromboembolism (VTE), a common condition in Western countries, is a cardiovascular disorder that arises due to haemostatic irregularities, which lead to thrombus generation inside veins. Even with successful treatment, the resulting disease spectrum of complications considerably affects the patient's quality of life, potentially leading to death. Cumulative data indicate that long non-coding RNAs (lncRNAs) may have a role in VTE pathogenesis. However, the clinical usefulness of these RNAs as biomarkers and potential therapeutic targets for VTE management is yet unclear. Thus, this article reviewed the emerging evidence on lncRNAs associated with VTE and with the activity of the coagulation system, which has a central role in disease pathogenesis. Until now, ten lncRNAs have been implicated in VTE pathogenesis, among which MALAT1 is the one with more evidence. Meanwhile, five lncRNAs have been reported to affect the expression of TFPI2, an important anticoagulant protein, but none with a described role in VTE development. More investigation in this field is needed as lncRNAs may help dissect VTE pathways, aiding in disease prediction, prevention and treatment.
Collapse
Affiliation(s)
- Inês Soares Marques
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Dep., Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; (I.S.M.); (V.T.); (B.V.N.); (I.N.R.M.)
- Faculty of Sciences of University of Porto (FCUP), 4169-007 Porto, Portugal
| | - Valéria Tavares
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Dep., Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; (I.S.M.); (V.T.); (B.V.N.); (I.N.R.M.)
- Faculty of Medicine of University of Porto (FMUP), 4200-072 Porto, Portugal
- Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Beatriz Vieira Neto
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Dep., Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; (I.S.M.); (V.T.); (B.V.N.); (I.N.R.M.)
- Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
| | - Inês N. R. Mota
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Dep., Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; (I.S.M.); (V.T.); (B.V.N.); (I.N.R.M.)
- Faculty of Sciences of University of Porto (FCUP), 4169-007 Porto, Portugal
| | - Deolinda Pereira
- Oncology Department, Portuguese Institute of Oncology of Porto (IPOP), 4200-072 Porto, Portugal;
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Dep., Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; (I.S.M.); (V.T.); (B.V.N.); (I.N.R.M.)
- Faculty of Medicine of University of Porto (FMUP), 4200-072 Porto, Portugal
- Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
- Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
| |
Collapse
|
24
|
Leung HM, Au SCL. Retinal Vein Occlusion after COVID-19 Vaccination-A Review. Vaccines (Basel) 2023; 11:1281. [PMID: 37631850 PMCID: PMC10459858 DOI: 10.3390/vaccines11081281] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 08/27/2023] Open
Abstract
Background Retinal vein occlusion (RVO) occurring after COVID-19 vaccination has been reported worldwide. Such a sight-threatening condition occurring after COVID-19 vaccination is a menace to ophthalmic health. This article reviews current evidence related to post-COVID-19 vaccination RVO. Method A total of 29 relevant articles identified on PubMed in January 2023 were selected for review. Observation All cases presented to ophthalmologists with visual loss shortly after COVID-19 vaccination. Mean and median age were both 58. No sex predominance was observed. RVO was diagnosed from findings on dilated fundal examination and ophthalmic imaging. AstraZeneca and BNT vaccines accounted for most cases. Vascular risk factors, e.g., diabetes mellitus and hypertension, were common. Most laboratory tests requested came back unremarkable. Most patients responded well to standard treatment, except those with ophthalmic comorbidities. Visual prognosis was excellent on short-term follow-up. Discussion The causality between RVO and COVID-19 vaccination is undeterminable because of the nature of articles, heterogenous reporting styles, contradicting laboratory findings and co-existing vascular risk factors. Vaccine-induced immune thrombotic thrombocytopenia, retinal vasculitis and homocysteinaemia were proposed to explain post-vaccination RVO. Large-scale studies have demonstrated that the incidence of RVO following COVID vaccination is very low. Nevertheless, the effects of boosters on retinal vasculature and ophthalmic health are still unclear. Conclusions The benefits of COVID-19 vaccination are believed to outweigh its ophthalmic risks. To ensure safe vaccination, the prior optimisation of comorbidities and post-vaccination monitoring are important. COVID-19 vaccines (including boosters) should be offered with reasonable confidence. Further studies are warranted to elucidate the ophthalmic impact of vaccines.
Collapse
Affiliation(s)
- Ho-Man Leung
- Hospital Authority, Ma Tau Wai 999077, Hong Kong
| | - Sunny Chi-Lik Au
- Department of Ophthalmology, Tung Wah Eastern Hospital, So Kon Po 999077, Hong Kong
| |
Collapse
|
25
|
Song J, Liu Y, Huang G. Predictive value of von Willebrand factor for venous thrombosis in patients with chronic heart failure complicated with atrial fibrillation after anticoagulant therapy. BMC Cardiovasc Disord 2023; 23:349. [PMID: 37442989 PMCID: PMC10347722 DOI: 10.1186/s12872-023-03167-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 03/02/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND We investigated the value of von Willebrand factor (vWF) in predicting venous thrombosis in patients with chronic heart failure complicated with atrial fibrillation after anticoagulation therapy. METHODS Totally, 126 patients with chronic heart failure complicated with atrial fibrillation who were treated with anticoagulant therapy and 60 healthy individuals were enrolled. One year after anticoagulant therapy, venous thrombosis occurred in 19 patients. Clinical data of patients were collected. The plasma vWF activity was detected and compared. The logistic regression analysis was used to analyze the influencing factors of vWF. ROC curve was used to evaluate the predictive value of plasma vWF. RESULTS Plasma vWF activity was significantly higher in patients with heart failure and atrial fibrillation than control subjects (P < 0.01). The vWF activity in patients with venous thrombosis was significantly higher than that in patients without venous thrombosis (P < 0.01). ROC curve analysis showed that the cut-off value of vWF activity for venous thrombosis within one year after anticoagulant therapy was 267.5%, and the AUC was 0.742 (95% CI: 0.764-0.921, P < 0.05). The sensitivity was 80.0%, and the specificity was 63.6%. Factors of diabetes, myocardial ischemia, old myocardial infarction, and lower extremity atherosclerosis, but not sex, age, coronary heart disease, hypertension, and cardiac function, had significant effect on vWF activity (P < 0.05). Logistic regression analysis showed that vWF activity was significantly related with atherosclerosis of lower limbs and old myocardial infarction, but not significantly related with diabetes and myocardial ischemia. The risk of venous thrombosis in patients with vWF activity greater than 267.5% was 10.667 times higher than that in patients with vWF activity less than 267.5% (P < 0.05). CONCLUSION The vWF activity greater than 267.5% has clinical predictive value for the risk of lower extremity venous thrombosis in patients with chronic heart failure complicated with atrial fibrillation within 1 year of anticoagulant therapy.
Collapse
Affiliation(s)
- Jinping Song
- Department of Clinical Laboratory, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91, Tianchi Road, Tianshan District, 830001 Urumqi, P.R. China
| | - Yuan Liu
- Department of Clinical Laboratory, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91, Tianchi Road, Tianshan District, 830001 Urumqi, P.R. China
| | - Guohong Huang
- Department of Clinical Laboratory, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91, Tianchi Road, Tianshan District, 830001 Urumqi, P.R. China
| |
Collapse
|
26
|
Prakash S, Bies J, Hassan M, Mares A, Didia SC. Portal vein thrombosis in cirrhosis: A literature review. Front Med (Lausanne) 2023; 10:1134801. [PMID: 37181351 PMCID: PMC10169608 DOI: 10.3389/fmed.2023.1134801] [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: 12/30/2022] [Accepted: 03/03/2023] [Indexed: 05/16/2023] Open
Abstract
Portal Vein Thrombosis (PVT), a common complication of advanced liver disease, is defined as an obstruction of the portal vein due to thrombus formation that can extend to the superior mesenteric and splenic veins. It was believed that PVT occurred predominantly due to prothrombotic potential. However, recent studies have shown that decreased blood flow related to portal hypertension appears to increase PVT risk as per Virchow's triad. It is well known that there is a higher incidence of PVTs in cirrhosis with a higher MELD and Child Pugh score. The controversy for management of PVTs in cirrhotics lies in the individualized assessment of risks versus benefits of anticoagulation, since these patients have a complex hemostatic profile with both bleeding and procoagulant propensities. In this review, we will systematically compile the etiology, pathophysiology, clinical features, and management of portal vein thrombosis in cirrhosis.
Collapse
Affiliation(s)
- Swathi Prakash
- Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States
| | - Jared Bies
- Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States
| | - Mariam Hassan
- Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States
| | - Adriana Mares
- Paul L. Foster School of Medicine, El Paso, TX, United States
| | - S. Claudia Didia
- Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States
| |
Collapse
|
27
|
Hellou T, Cohen O, Avigdor A, Amitai I, Shimoni A, Misgav M, Canaani J. The occurrence of thrombosis during intensive chemotherapy treatment for acute myeloid leukemia patients does not impact on long-term survival. Ann Hematol 2023; 102:1037-1043. [PMID: 36905445 DOI: 10.1007/s00277-023-05158-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/02/2023] [Indexed: 03/12/2023]
Abstract
Venous thromboembolism (VTE) is frequently seen in acute myeloid leukemia (AML) patients and presents a significant clinical challenge. The association of VTE during intensive chemotherapy with risk models such as the Medical Research Council (MRC) cytogenetic-based assessment and the European LeukemiaNet (ELN) 2017 molecular risk model have not been rigorously evaluated. Additionally, there is a paucity of data pertaining to the long-term prognostic impact of VTE in AML patients. We performed an analysis of baseline parameters of AML patients diagnosed with VTE during intensive chemotherapy and compared them with patients without VTE. The analyzed cohort consisted of 335 newly diagnosed AML patients with a median age of 55 years. Thirty-five patients (11%) were classified as MRC favorable risk, 219 (66%) patients as intermediate risk, 58 patients (17%) as adverse risk. Per ELN 2017, 132 patients (40%) had favorable risk disease, 122 patients (36%) intermediate risk, and 80 patients (24%) had adverse risk. VTE was seen in 33 patients (9.9%), occurring mostly during induction (70%), and required catheter removal in 9 patients (28%). Baseline clinical, laboratory, molecular, and ELN 2017 parameters were not significantly different groups. However, MRC intermediate-risk group patients were significantly more likely to experience thrombosis compared to favorable risk and adverse risk patients (12.8% versus 5.7% and 1.7%, respectively; p = 0.049). Median overall survival was not significantly impacted by the diagnosis of thrombosis (3.7 years versus 2.2 years; p = 0.47). VTE is tightly associated with temporal and cytogenetic parameters in AML but does not significantly impact on long-term outcomes.
Collapse
Affiliation(s)
- Tamer Hellou
- Hematology Division, Sheba Medical Center, Faculty of Medicine, Tel Aviv University, 52621, Tel-Hashomer, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Omri Cohen
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- National Hemophilia Center, Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel-Hashomer, Israel
| | - Abraham Avigdor
- Hematology Division, Sheba Medical Center, Faculty of Medicine, Tel Aviv University, 52621, Tel-Hashomer, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Irina Amitai
- Hematology Division, Sheba Medical Center, Faculty of Medicine, Tel Aviv University, 52621, Tel-Hashomer, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avichai Shimoni
- Hematology Division, Sheba Medical Center, Faculty of Medicine, Tel Aviv University, 52621, Tel-Hashomer, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mudi Misgav
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- National Hemophilia Center, Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel-Hashomer, Israel
| | - Jonathan Canaani
- Hematology Division, Sheba Medical Center, Faculty of Medicine, Tel Aviv University, 52621, Tel-Hashomer, Israel.
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
28
|
DeRoo E, Zhou T, Yang H, Stranz A, Henke P, Liu B. A vein wall cell atlas of murine venous thrombosis determined by single-cell RNA sequencing. Commun Biol 2023; 6:130. [PMID: 36721040 PMCID: PMC9889765 DOI: 10.1038/s42003-023-04492-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 01/16/2023] [Indexed: 02/02/2023] Open
Abstract
Deep vein thrombosis (DVT) is a common clinical problem, but its cellular and molecular mechanisms remain incompletely understood. In this study, we performed single-cell RNA sequencing on mouse inferior vena cava (IVC) 24 h after thrombus-inducing IVC ligation or sham operation. 9 cell types composed of multiple subpopulations were identified. Notable transcriptomic changes induced by DVT included a marked inflammatory response, elevated hypoxia, and globally reduced myogenesis. Analysis of individual cell populations revealed increased inflammation and reduced extracellular matrix production across smooth muscle cells and fibroblasts, juxtaposed against an early phenotypic shift in smooth muscle cell populations away from a contractile state. By characterizing the transcriptomic changes in the vein wall during acute venous thrombosis at the single-cell level, this work provides novel insights into early pathological events in the vein wall that may potentiate thrombus formation and result in long term adverse venous remodeling.
Collapse
Affiliation(s)
- Elise DeRoo
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Ting Zhou
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Huan Yang
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Amelia Stranz
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Peter Henke
- Department of Surgery, Division of Vascular Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Bo Liu
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Cellular and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
29
|
Siwiec-Kozlik A, Kuszmiersz P, Kasper L, Frolow M, Kozlik-Siwiec P, Iwaniec T, Kosalka-Wegiel J, Zareba L, Sladek K, Bazan JG, Bazan-Socha S, Dropinski J. Prothrombotic state, endothelial injury, and echocardiographic changes in non-active sarcoidosis patients. Sci Rep 2022; 12:21291. [PMID: 36494464 PMCID: PMC9734106 DOI: 10.1038/s41598-022-25580-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
Sarcoidosis is a multisystem inflammatory granulomatous disease of unknown cause that most commonly affects lungs and lymph nodes, with frequent yet asymptomatic cardiac involvement. The epidemiologically associated cardiovascular risk suggests an underlying prothrombotic state and endothelial dysfunction, currently understudied in the available literature. Therefore, we aimed to investigate prothrombotic plasma properties together with selected echocardiographic and laboratory biomarkers of cardiovascular injury in that disease. N = 53 patients with pulmonary sarcoidosis in clinical remission and N = 66 matched controls were assessed for inflammatory and endothelial injury biomarkers, plasma thrombin generation profile, and echocardiographic and lung function parameters. Sarcoidosis cases had impaired systolic and diastolic left ventricular function, higher concentrations of inflammatory markers, D-dimer and factor VIII activity compared to the controls. The coexistence of extrapulmonary disease was associated with elevated circulating vascular cell adhesion molecule 1, while cases with hypercalcemia had higher thrombomodulin concentration. Sarcoidosis was characterized by the unfavorably altered thrombin generation profile, reflected by the 16% higher endogenous thrombin potential (ETP), 24% increased peak thrombin concentration, and 12% shorter time to thrombin peak in comparison to the control group. ETP was higher in cases with proxies of pulmonary restriction, extrapulmonary-extracutaneous manifestation, and need for corticosteroids use. Despite the clinical remission, sarcoidosis is related to prothrombotic plasma properties and signs of endothelial injury, likely contributing to the higher risk of cardiovascular events. In addition, subclinical cardiac involvement may play an additional role, although further clinical and experimental studies are needed to verify these findings.
Collapse
Affiliation(s)
- Andzelika Siwiec-Kozlik
- grid.5522.00000 0001 2162 9631Department of Rheumatology and Immunology, Jagiellonian University Medical College, Cracow, Poland ,grid.412700.00000 0001 1216 0093Rheumatology and Immunology Clinical Department, University Hospital, Cracow, Poland
| | - Piotr Kuszmiersz
- grid.5522.00000 0001 2162 9631Department of Rheumatology and Immunology, Jagiellonian University Medical College, Cracow, Poland ,grid.412700.00000 0001 1216 0093Rheumatology and Immunology Clinical Department, University Hospital, Cracow, Poland
| | - Lukasz Kasper
- grid.5522.00000 0001 2162 9631Department of Internal Medicine, Jagiellonian University Medical College, Cracow, Poland ,grid.412700.00000 0001 1216 0093Pulmonology and Allergology Clinical Department, University Hospital, Cracow, Poland
| | - Marzena Frolow
- grid.5522.00000 0001 2162 9631Department of Internal Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Pawel Kozlik-Siwiec
- grid.5522.00000 0001 2162 9631Department of Internal Medicine, Jagiellonian University Medical College, Cracow, Poland ,grid.412700.00000 0001 1216 0093Hematology Clinical Department, University Hospital, Cracow, Poland
| | - Teresa Iwaniec
- grid.5522.00000 0001 2162 9631Department of Hematology, Jagiellonian University Medical College, Cracow, Poland
| | - Joanna Kosalka-Wegiel
- grid.5522.00000 0001 2162 9631Department of Rheumatology and Immunology, Jagiellonian University Medical College, Cracow, Poland ,grid.412700.00000 0001 1216 0093Rheumatology and Immunology Clinical Department, University Hospital, Cracow, Poland
| | - Lech Zareba
- grid.13856.390000 0001 2154 3176Institute of Computer Science, College of Natural Sciences, University of Rzeszow, Rzeszow, Poland
| | - Krzysztof Sladek
- grid.5522.00000 0001 2162 9631Department of Internal Medicine, Jagiellonian University Medical College, Cracow, Poland ,grid.412700.00000 0001 1216 0093Pulmonology and Allergology Clinical Department, University Hospital, Cracow, Poland
| | - Jan G. Bazan
- grid.13856.390000 0001 2154 3176Institute of Computer Science, College of Natural Sciences, University of Rzeszow, Rzeszow, Poland
| | - Stanislawa Bazan-Socha
- grid.5522.00000 0001 2162 9631Department of Internal Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Jerzy Dropinski
- grid.5522.00000 0001 2162 9631Department of Internal Medicine, Jagiellonian University Medical College, Cracow, Poland
| |
Collapse
|
30
|
da Silva FC, de Araújo BJ, Cordeiro CS, Arruda VM, Faria BQ, Guerra JFDC, Araújo TGD, Fürstenau CR. Endothelial dysfunction due to the inhibition of the synthesis of nitric oxide: Proposal and characterization of an in vitro cellular model. Front Physiol 2022; 13:978378. [PMID: 36467706 PMCID: PMC9714775 DOI: 10.3389/fphys.2022.978378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 11/02/2022] [Indexed: 08/13/2023] Open
Abstract
The vascular endothelium plays a pivotal role in the maintenance of vascular homeostasis, mediated by vasoactive molecules produced by endothelial cells. The balance between vasoconstrictor and vasodilator biomolecules is what guarantees this equilibrium. Therefore, an increase in the bioavailability of vasoconstrictors along with a reduction in vasodilators may indicate a condition known as endothelial dysfunction. Endothelial dysfunction is marked by an inflammatory process and reduced activity of vasoprotective enzymes, being characterized by some factors like the reduction of the bioavailability of nitric oxide (NO) and increase in the production of reactive oxygen species (ROS), pro-inflammatory and vasoconstrictor molecules. This condition is a predictive marker of several cardiovascular diseases (e.g., atherosclerosis, hypertension, and diabetes). Research is affected by the scarcity of suitable in vitro models that simulate endothelial dysfunction. The goal of this study was to induce an in vitro condition to mimic endothelial dysfunction by inhibiting NO synthesis in cells. Thymus-derived endothelial cells (tEnd.1) were treated with different concentrations of L-NAME (from 1 to 1,000 μM) for different times (12, 24, 48, 72, 96, and 120 h without and with retreatment every 24 h). Cell viability, nitrite concentration, p22phox, NOX2, NOX4, IL-6, and ACE genes expression and lipid peroxidation were evaluated. The results indicate that the treatment with 100 μM L-NAME for 72 h without retreatment reduced NO concentration and NOX4 gene expression while increasing ACE expression, thus mimicking reduced vascular protection and possibly increased vasoconstriction. On the other hand, treatment with 100 μM L-NAME for 96 h with retreatment reduced the concentration of NO and the expression of the p22phox gene while increasing the expression of the IL-6 and ACE genes, mimicking the increase in inflammation and vasoconstriction parameters. Based on these results, we thus propose that both 100 μM L-NAME for 72 h without retreatment and 100 μM L-NAME for 96 h with retreatment may be used as models for in vitro endothelial dysfunction according to the purpose of the study to be conducted.
Collapse
Affiliation(s)
- Fernanda Cardoso da Silva
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
- Laboratory of Vascular Biochemistry, Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil
| | - Bruna Juber de Araújo
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
- Laboratory of Vascular Biochemistry, Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil
| | - Carina Santos Cordeiro
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
| | - Vinícius Marques Arruda
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
| | - Bruno Quintanilha Faria
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
| | - Joyce Ferreira Da Costa Guerra
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
| | - Thaise Gonçalves De Araújo
- Animal Cell Culture Laboratory, Institute of Biotechnology, Federal University of Uberlândia, Patos de Minas, MG, Brazil
| | - Cristina Ribas Fürstenau
- Laboratory of Vascular Biochemistry, Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil
| |
Collapse
|
31
|
Luu B, Ruderman S, Nance R, Delaney JAC, Ma J, Hahn A, Heckbert SR, Budoff MJ, Crothers K, Mathews WC, Christopolous K, Hunt PW, Eron J, Moore R, Keruly J, Lober WB, Burkholder GA, Willig A, Chander G, McCaul ME, Cropsey K, O'Cleirigh C, Peter I, Feinstein M, Tsui JI, Lindstroem S, Saag M, Kitahata MM, Crane HM, Drumright LN, Whitney BM. Tobacco smoking and binge alcohol use are associated with incident venous thromboembolism in an HIV cohort. HIV Med 2022; 23:1051-1060. [PMID: 35343038 PMCID: PMC9515244 DOI: 10.1111/hiv.13309] [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: 06/22/2021] [Revised: 01/27/2022] [Accepted: 03/14/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND People with HIV (PWH) are at increased risk of cardiovascular comorbidities and substance use is a potential predisposing factor. We evaluated associations of tobacco smoking and alcohol use with venous thromboembolism (VTE) in PWH. METHODS We assessed incident, centrally adjudicated VTE among 12 957 PWH within the Centers for AIDS Research Network of Integrated Clinical Systems (CNICS) cohort between January 2009 and December 2018. Using separate Cox proportional hazards models, we evaluated associations of time-updated alcohol and cigarette use with VTE, adjusting for demographic and clinical characteristics. Smoking was evaluated as pack-years and never, former, or current use with current cigarettes per day. Alcohol use was parameterized using categorical and continuous alcohol use score, frequency of use, and binge frequency. RESULTS During a median of 3.6 years of follow-up, 213 PWH developed a VTE. One-third of PWH reported binge drinking and 40% reported currently smoking. In adjusted analyses, risk of VTE was increased among both current (HR: 1.44, 95% CI: 1.02-2.03) and former (HR: 1.44, 95% CI: 0.99-2.07) smokers compared to PWH who never smoked. Additionally, total pack-years among ever-smokers (HR: 1.10 per 5 pack-years; 95% CI: 1.03-1.18) was associated with incident VTE in a dose-dependent manner. Frequency of binge drinking was associated with incident VTE (HR: 1.30 per 7 days/month, 95% CI: 1.11-1.52); however, alcohol use frequency was not. Severity of alcohol use was not significantly associated with VTE. CONCLUSIONS Current smoking and pack-year smoking history were dose-dependently associated with incident VTE among PWH in CNICS. Binge drinking was also associated with VTE. Interventions for smoking and binge drinking may decrease VTE risk among PWH.
Collapse
Affiliation(s)
- Brandon Luu
- Northern Ontario School of Medicine, Thunder Bay, Ontario, Canada
| | | | - Robin Nance
- University of Washington, Seattle, Washington, USA
| | - Joseph A C Delaney
- University of Washington, Seattle, Washington, USA
- University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jimmy Ma
- University of Washington, Seattle, Washington, USA
| | - Andrew Hahn
- University of Washington, Seattle, Washington, USA
| | | | | | | | | | | | - Peter W Hunt
- University of California, San Francisco, California, USA
| | - Joseph Eron
- University of North Carolina, Chapel Hill, North Carolina, USA
| | | | | | | | | | - Amanda Willig
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | - Karen Cropsey
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Inga Peter
- Mount Sinai Hospital, New York, New York, USA
| | | | | | | | - Michael Saag
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | | | | |
Collapse
|
32
|
Immunothrombosis and the Role of Platelets in Venous Thromboembolic Diseases. Int J Mol Sci 2022; 23:ijms232113176. [PMID: 36361963 PMCID: PMC9656618 DOI: 10.3390/ijms232113176] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 12/05/2022] Open
Abstract
Venous thromboembolism (VTE) is the third leading cardiovascular cause of death and is conventionally treated with anticoagulants that directly antagonize coagulation. However, recent data have demonstrated that also platelets play a crucial role in VTE pathophysiology. In the current review, we outline how platelets are involved during all stages of experimental venous thrombosis. Platelets mediate initiation of the disease by attaching to the vessel wall upon which they mediate leukocyte recruitment. This process is referred to as immunothrombosis, and within this novel concept inflammatory cells such as leukocytes and platelets directly drive the progression of VTE. In addition to their involvement in immunothrombosis, activated platelets can directly drive venous thrombosis by supporting coagulation and secreting procoagulant factors. Furthermore, fibrinolysis and vessel resolution are (partly) mediated by platelets. Finally, we summarize how conventional antiplatelet therapy can prevent experimental venous thrombosis and impacts (recurrent) VTE in humans.
Collapse
|
33
|
Akrivou D, Perlepe G, Kirgou P, Gourgoulianis KI, Malli F. Pathophysiological Aspects of Aging in Venous Thromboembolism: An Update. Medicina (B Aires) 2022; 58:medicina58081078. [PMID: 36013544 PMCID: PMC9415158 DOI: 10.3390/medicina58081078] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/03/2022] [Accepted: 08/07/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of this review is to highlight all the factors that associate venous thromboembolism (VTE) with aging. Elderly people are characterized by a higher incidence of thrombosis taking into account the co-existing comorbidities, complications and fatality that arise. Based on the Virchow triad, pathophysiological aspects of venous stasis, endothelium injury and hypercoagulability in elderly people (≥65 years) are described in detail. More precisely, venous wall structure, nitric oxide (NO) and endothelin-1 expression are impaired in this age group. Furthermore, an increase in high-molecular-weight kininogen (HMWK), prekallikrein, factors V, VII, VIII, IX and XI, clot lysis time (CLT) and von Willebrand factor (vWF) is observed. Age-dependent platelet dysfunction and changes in anticoagulant factors are also illustrated. A “low-grade inflammation stage” is delineated as a possible risk factor for thrombosis in the elderly. Consequently, clinical implications for frail elderly people related to diagnosis, treatment, bleeding danger and VTE recurrence emerge. We conclude that aging is an acquired thrombotic factor closely related to pathophysiological changes.
Collapse
Affiliation(s)
- Dimitra Akrivou
- Respiratory Medicine Department, Faculty of Medicine, University of Thessaly, 41300 Larissa, Greece
| | - Garifallia Perlepe
- Respiratory Medicine Department, Faculty of Medicine, University of Thessaly, 41300 Larissa, Greece
| | - Paraskevi Kirgou
- Respiratory Medicine Department, Faculty of Medicine, University of Thessaly, 41300 Larissa, Greece
| | | | - Foteini Malli
- Respiratory Medicine Department, Faculty of Medicine, University of Thessaly, 41300 Larissa, Greece
- Respiratory Disorders Lab, Faculty of Nursing, University of Thessaly, 41300 Larissa, Greece
- Respiratory Medicine Department, University Hospital of Larissa, 41223 Larissa, Greece
- Correspondence: ; Tel.: +30-2410684612
| |
Collapse
|
34
|
Predicting the risk of postsplenectomy thrombosis in patients with portal hypertension using computational hemodynamics models: A proof-of-concept study. Clin Biomech (Bristol, Avon) 2022; 98:105717. [PMID: 35834965 DOI: 10.1016/j.clinbiomech.2022.105717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/05/2022] [Accepted: 07/06/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND The high incidence of thrombosis in the portal venous system following splenectomy (a frequently adopted surgery for treating portal hypertension in patients with splenomegaly and hypersplenism) is a critical clinical issue. The aim of this study was to address whether quantification of postsplenectomy hemodynamics has potential value for assessing the risk of postsplenectomy thrombosis. METHODS Computational models were constructed for three portal hypertensive patients treated with splenectomy based on their preoperative clinical data to quantify hemodynamics in the portal venous system before and after splenectomy, respectively. Each patient was followed up for three or five months after surgery and examined with CT to screen potential thrombosis. FINDINGS The area ratio of wall regions exposed to low wall shear stress was small before splenectomy in all patients, which increased markedly after splenectomy and exhibited enlarged inter-patient differences. The largest area ratio of low wall shear stress and most severe flow stagnation after splenectomy were predicted for the patient suffering from postsplenectomy thrombosis, with the wall regions exposed to low wall shear stress corresponding well with the CT-detected distribution of thrombus. Further analyses revealed that postoperative hemodynamic characteristics were considerably influenced by the anatomorphological features of the portal venous system. INTERPRETATION Postoperative hemodynamic conditions in the portal venous system are highly patient-specific and have a potential link to postsplenectomy thrombosis, which indicates that patient-specific hemodynamic studies may serve as a complement to routine clinical assessments for refining risk stratification and postoperative patient management.
Collapse
|
35
|
Li Y, Ge J, Ma K, Kong J. Epigallocatechin-3-gallate exerts protective effect on epithelial function via PI3K/AKT signaling in thrombosis. Microvasc Res 2022; 144:104408. [PMID: 35878868 DOI: 10.1016/j.mvr.2022.104408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/27/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Venous thrombosis (VT) is one of the most frequent cardiovascular diseases, which seriously endangers people's health. Recently, the protective role of epigallocatechin-3-gallate (EGCG) against multiple cardiovascular diseases has been well studied. Nevertheless, whether EGCG is implicated in the progression of VT is still unclear. METHODS Rat models of VT were established by inferior vena cava (IVC) ligation. Histological characterization of the IVC tissues was examined by hematoxylin-eosin (H&E) staining. TUNEL assay was utilized to detect cell apoptosis in IVC tissues. The concentrations of the oxidative stress biomarkers, malondialdehyde (MDA) and superoxide dismutase (SOD), were estimated by corresponding kits. In addition, the levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-8 in rat plasma were estimated by ELISA. Further, the expression levels of apoptosis markers (Bax, Bcl-2, and Cleaved-caspase 3) as well as key molecules p-PI3K and p-AKT in phosphoinositide 3-kinase (PI3K)/AKT signaling pathway were assessed by western blot. RESULTS Compared to the sham group, the model group showed obvious thrombus formation in IVC tissues, while the EGCG treatment significantly repressed thrombosis. EGCG inhibited cell apoptosis in IVC tissues of VT rat models. The decreased SOD concentration and increased MDA concentration in the plasma of VT rats were reversed by EGCG treatment. Additionally, the elevated levels of TNF-α, IL-6 and IL-8 in the plasma of VT rats can be partially reduced by the treatment of EGCG. Finally, we also found that EGCG reduced the levels of phosphorylated (p)-PI3K and p-AKT in IVC tissues of VT rat models, indicating that the hyperactivation of the PI3K/AKT signaling pathway was inhibited by EGCG. CONCLUSION This study proves that EGCG alleviates thrombosis, cell apoptosis, inflammatory response, and oxidative stress injury in VT by inactivating PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Yan Li
- Department of Vascular and Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Jingping Ge
- Department of Vascular and Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Ke Ma
- Department of Acupuncture, Qinhuai District Hospital of Traditional Chinese Medicine, Nanjing 210000, Jiangsu, China
| | - Jie Kong
- Department of Vascular and Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China.
| |
Collapse
|
36
|
Tetramethylpyrazine Protects Endothelial Injury and Antithrombosis via Antioxidant and Antiapoptosis in HUVECs and Zebrafish. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2232365. [PMID: 35898617 PMCID: PMC9313999 DOI: 10.1155/2022/2232365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022]
Abstract
Chuanxiong Rhizoma, the dried rhizome of Ligusticum chuanxiong Hort., is a commonly used drug for promoting blood circulation and dissipating congestion. Tetramethylpyrazine (TMP), the main active ingredient of Ligusticum chuanxiong, has significant antioxidant, anti-inflammatory, and vascular protective effects. However, the protective properties and underlying mechanisms of TMP against endothelial injury-induced insufficient angiogenesis and thrombosis have not been elucidated. Therefore, we aimed to explore the protective effects of TMP on endothelial injury and its antithrombotic effects and study the mechanism. In vitro experiments showed that TMP could alleviate hydrogen peroxide– (H2O2–) induced endothelial injury of human umbilical vein endothelial cells (HUVECs) and the protective mechanism might be related to the regulation of MAPK signaling pathway, and its antioxidative and antiapoptotic effects. In vivo experiments showed that TMP restored PTK787-induced damage to intersegmental vessels (ISVs) in Tg(fli-1: EGFP)y1 transgenic (Flik) zebrafish larvae. Similarly, adrenalin hydrochloride– (AH–) induced reactive oxygen species (ROS) production and thrombosis in AB strain zebrafish were inhibited by TMP. RT-qPCR assay proved that TMP could inhibit the expression of fga, fgb, fgg, f7, and von Willebrand factor (vWF) mRNA to exert an antithrombotic effect. Our findings suggest that TMP can contribute to endothelial injury protection and antithrombosis by modulating MAPK signaling and attenuating oxidative stress and antiapoptosis.
Collapse
|
37
|
Wang H, Lin S, Yang Y, Zhao M, Li X, Zhang L. Significant role of long non-coding RNA MALAT1 in deep vein thrombosis via the regulation of vascular endothelial cell physiology through the microRNA-383-5p/BCL2L11 axis. Bioengineered 2022; 13:13728-13738. [PMID: 35706417 PMCID: PMC9276002 DOI: 10.1080/21655979.2022.2080412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Deep vein thrombosis (DVT) is a vascular disease. The long non-coding RNA (lncRNA), metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), is positively expressed in DVT tissues, and regulates the biological behavior of endothelial progenitor cells. Here, we explored whether MALAT1 affected the physiology of human vascular endothelial cells (HUVECs) and analyzed its underlying mechanism. To overexpress/silence the expression of MALAT1 in HUVECs, MALAT1-plasmid/MALAT1-small interfering RNA (siRNA) was used. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide and flow cytometry analyses were performed to observe the cell viability and apoptosis. Reverse transcription-quantitative polymerase chain reaction and western blotting were used to determine the apoptosis-related protein and gene expression levels. We used Starbase software to predict the associations among MALAT1, microRNA (miR)-383-5p, and BCL2-like 11 (BCL2L11). Luciferase reporter assay was used to validate their relationship. Compared to the control vector group, MALAT1-plasmid suppressed the viability and induced apoptosis of HUVECs, while improving Bcl-2-associated X protein (Bax) expression and decreasing Bcl-2 expression. There was an interaction between MALAT1 and miR-383-5p. Compared to the control siRNA group, MALAT1-siRNA increased the cell viability, reduced cell apoptosis, upregulated Bcl-2 expression, and suppressed Bax expression. These changes were reversed by the miR-383-5p inhibitor. Additionally, we verified that BCL2L11 is a target of miR-383-5p. miR-383-5p improved the cell proliferation, while decreasing cell apoptosis in HUVECs by targeting BCL2L11. Therefore, the lncRNA-MALAT1/miR-383-5p/BCL2L11 axis may be effective for DVT treatment.
Collapse
Affiliation(s)
- Hecheng Wang
- Department of Academic Affairs, The Third Affiliated Hospital of Qiqihar Medical College, Qiqihar 161000, China
| | - Shusen Lin
- Department of Vascular Surgery, The Third Affiliated Hospital of Qiqihar Medical College, Qiqihar 161000, China
| | - Yujie Yang
- Department of Academic Affairs, The Third Affiliated Hospital of Qiqihar Medical College, Qiqihar 161000, China
| | - Mingyu Zhao
- Department of Vascular Surgery, The Third Affiliated Hospital of Qiqihar Medical College, Qiqihar 161000, China
| | - Xichun Li
- Department of Vascular Surgery, The Third Affiliated Hospital of Qiqihar Medical College, Qiqihar 161000, China
| | - Lanli Zhang
- Department of Ultrasound, The Third Affiliated Hospital of Qiqihar Medical College, Qiqihar 161000, Chin
| |
Collapse
|
38
|
Targeting vascular inflammation through emerging methods and drug carriers. Adv Drug Deliv Rev 2022; 184:114180. [PMID: 35271986 PMCID: PMC9035126 DOI: 10.1016/j.addr.2022.114180] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 02/18/2022] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
Abstract
Acute inflammation is a common dangerous component of pathogenesis of many prevalent conditions with high morbidity and mortality including sepsis, thrombosis, acute respiratory distress syndrome (ARDS), COVID-19, myocardial and cerebral ischemia-reperfusion, infection, and trauma. Inflammatory changes of the vasculature and blood mediate the course and outcome of the pathology in the tissue site of insult, remote organs and systemically. Endothelial cells lining the luminal surface of the vasculature play the key regulatory functions in the body, distinct under normal vs. pathological conditions. In theory, pharmacological interventions in the endothelial cells might enable therapeutic correction of the overzealous damaging pro-inflammatory and pro-thrombotic changes in the vasculature. However, current agents and drug delivery systems (DDS) have inadequate pharmacokinetics and lack the spatiotemporal precision of vascular delivery in the context of acute inflammation. To attain this level of precision, many groups design DDS targeted to specific endothelial surface determinants. These DDS are able to provide specificity for desired tissues, organs, cells, and sub-cellular compartments needed for a particular intervention. We provide a brief overview of endothelial determinants, design of DDS targeted to these molecules, their performance in experimental models with focus on animal studies and appraisal of emerging new approaches. Particular attention is paid to challenges and perspectives of targeted therapeutics and nanomedicine for advanced management of acute inflammation.
Collapse
|
39
|
Navarrete S, Solar C, Tapia R, Pereira J, Fuentes E, Palomo I. Pathophysiology of deep vein thrombosis. Clin Exp Med 2022:10.1007/s10238-022-00829-w. [PMID: 35471714 DOI: 10.1007/s10238-022-00829-w] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 03/31/2022] [Indexed: 12/29/2022]
Abstract
Deep venous thrombosis is a frequent, multifactorial disease and a leading cause of morbidity and mortality. Most of the time deep venous thrombosis is triggered by the interaction between acquired risk factors, such as hip fracture, pregnancy, and immobility, and hereditary risk factors such as thrombophilias. The mechanisms underlying deep venous thrombosis are not fully elucidated; however, in recent years, important advances have shed light on the role of venous flow, endothelium, platelets, leukocytes, and the interaction between inflammation and hemostasis. It has been described that the alteration of venous blood flow produces endothelial activation, favoring the adhesion of platelets and leukocytes, which, through tissue factor expression and neutrophil extracellular traps formation, contribute to the activation of coagulation, trapping more cells, such as red blood cells. Thus, the concerted interaction of these phenomena allows the formation and growth of the thrombus. In this work, the main mechanisms involved in the pathophysiology of deep vein thrombosis will be described.
Collapse
Affiliation(s)
- Simón Navarrete
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Av. Lircay s/n, 3460000, Talca, Chile
| | - Carla Solar
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Av. Lircay s/n, 3460000, Talca, Chile
| | | | - Jaime Pereira
- Department of Hematology-Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Av. Lircay s/n, 3460000, Talca, Chile
| | - Iván Palomo
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Av. Lircay s/n, 3460000, Talca, Chile.
| |
Collapse
|
40
|
Raj R, Paturi A, Ahmed MA, Thomas SE, Gorantla VR. Obstructive Sleep Apnea as a Risk Factor for Venous Thromboembolism: A Systematic Review. Cureus 2022; 14:e22729. [PMID: 35371730 PMCID: PMC8971089 DOI: 10.7759/cureus.22729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2022] [Indexed: 12/14/2022] Open
Abstract
Obstructive sleep apnea (OSA), is a prevalent condition characterized by repeated episodes of pharyngeal airway obstruction resulting in hypopnea and apnea episodes during sleep leading to nightly awakenings. OSA is a major contributor to the healthcare burden worldwide due to its high cardiovascular morbidity and mortality. There is growing evidence to support a pathophysiological link between OSA and venous thromboembolism (VTE). The pro-inflammatory state along with intermittent hypoxia that is invoked in OSA is associated with blood hypercoagulability, venous stasis, and endothelial dysfunction leading to deep vein thrombosis (DVT) and pulmonary embolism (PE). In this systematic review, we aim to analyze and assess the available literature on OSA and VTE (or DVT/PE) to determine whether OSA is an independent risk factor for VTE.
Collapse
|
41
|
Turton HA, Pickworth J, Paterson GG, Lawrie A, Baillie JK, Thompson AAR. Soluble P-Selectin and von Willebrand Factor Rise in Healthy Volunteers Following Non-exertional Ascent to High Altitude. Front Physiol 2022; 13:825819. [PMID: 35250627 PMCID: PMC8889065 DOI: 10.3389/fphys.2022.825819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Reduced oxygen tensions experienced at high altitudes are thought to predispose to thrombosis, yet there are few studies linking hypoxia, platelet activation, and thrombosis. Reports of platelet phenotypes in hypoxia are inconsistent, perhaps due to differing degrees of hypoxia experienced and the duration of exposure. This study aimed to investigate the relationship between soluble P-selectin, a marker of platelet activation, and von Willebrand factor (vWF) on exposure to hypoxia. We measured plasma concentrations of P-selectin and vWF in sixteen healthy volunteers before, during and after the APEX 2 expedition. APEX 2 consisted of a non-exertional ascent to 5,200 m, followed by 7 consecutive days at high altitude. We showed that high altitude significantly increased mean plasma P-selectin and vWF compared to pre-expedition levels. Both plasma marker levels returned to baseline post-expedition. We found a strong positive correlation between vWF and P-selectin, but no association between P-selectin and platelet count. Our results are consistent with previous work showing evidence of platelet activation at high altitude and demonstrate that the rise in P-selectin is not simply due to an increase in platelet count. As vWF and P-selectin could be derived from either platelets or endothelial cells, further work assessing more specific markers of endothelial activation is proposed to provide insight into the source of these potential pro-thrombotic biomarkers at altitude.
Collapse
Affiliation(s)
- Helena A. Turton
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Josephine Pickworth
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Gordon G. Paterson
- Apex (Altitude Physiology Expeditions, SC030345), Edinburgh, United Kingdom
- Centre for Primary Care and Public Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - J. Kenneth Baillie
- Apex (Altitude Physiology Expeditions, SC030345), Edinburgh, United Kingdom
- Department of Anaesthesia, Critical Care and Pain Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - A. A. Roger Thompson
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
- Apex (Altitude Physiology Expeditions, SC030345), Edinburgh, United Kingdom
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| |
Collapse
|
42
|
The Pathophysiology of Portal Vein Thrombosis in Cirrhosis: Getting Deeper into Virchow's Triad. J Clin Med 2022; 11:jcm11030800. [PMID: 35160251 PMCID: PMC8837039 DOI: 10.3390/jcm11030800] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/21/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023] Open
Abstract
Portal vein thrombosis (PVT) is a common complication among patients with cirrhosis. However, its pathophysiology is not well established and there are currently very few predictive factors, none of which are actually useful, from a clinical perspective. The contribution of each of the vertices of Virchow’s triad, e.g., blood hypercoagulability, blood flow, and portal vein endothelial damage in the development of PVT is not clear. In this review, we aim to recapitulate the latest studies on the field of PVT development in order to understand its mechanisms and discuss some of the future directions in the study of this important complication of cirrhosis.
Collapse
|
43
|
Dardi P, Perazza LR, Couto GK, Campos GP, Capettini LDSA, Rossoni LV. Vena cava presents endothelial dysfunction prior to thoracic aorta in heart failure: the pivotal role of nNOS uncoupling/oxidative stress. Clin Sci (Lond) 2021; 135:2625-2641. [PMID: 34783347 DOI: 10.1042/cs20210810] [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: 08/11/2021] [Revised: 11/05/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022]
Abstract
Arterial endothelial dysfunction has been extensively studied in heart failure (HF). However, little is known about the adjustments shown by the venous system in this condition. Considering that inferior vena cava (VC) tone could influence cardiac performance and HF prognosis, the aim of the present study was to assess the VC and thoracic aorta (TA) endothelial function of HF-post-myocardial infarction (MI) rats, comparing both endothelial responses and signaling pathways developed. Vascular reactivity of TA and VC from HF post-MI and sham operated (SO) rats was assessed with a wire myograph, 4 weeks after coronary artery occlusion surgery. Nitric oxide (NO), H2O2 production and oxidative stress were evaluated in situ with fluorescent probes, while protein expression and dimer/monomer ratio was assessed by Western blot. VC from HF rats presented endothelial dysfunction, while TA exhibited higher acetylcholine (ACh)-induced vasodilation when compared with vessels from SO rats. TA exhibited increased ACh-induced NO production due to a higher coupling of endothelial and neuronal NO synthases isoforms (eNOS, nNOS), and enhanced expression of antioxidant enzymes. These adjustments, however, were absent in VC of HF post-MI rats, which exhibited uncoupled nNOS, oxidative stress and higher H2O2 bioavailability. Altogether, the present study suggests a differential regulation of endothelial function between VC and TA of HF post-MI rats, most likely due to nNOS uncoupling and compromised antioxidant defense.
Collapse
Affiliation(s)
- Patrizia Dardi
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Laís Rossi Perazza
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Gisele Kruger Couto
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Gianne Paul Campos
- Department of Pharmacology, Institute of Biological Science, University of Minas Gerais, Minas Gerais, Brazil
| | | | - Luciana Venturini Rossoni
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
44
|
Hegner P, Lebek S, Maier LS, Arzt M, Wagner S. The Effect of Gender and Sex Hormones on Cardiovascular Disease, Heart Failure, Diabetes, and Atrial Fibrillation in Sleep Apnea. Front Physiol 2021; 12:741896. [PMID: 34744785 PMCID: PMC8564381 DOI: 10.3389/fphys.2021.741896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/27/2021] [Indexed: 12/29/2022] Open
Abstract
Sleep apnea is a highly prevalent disorder with increasing impact on healthcare systems worldwide. Previous studies have been conducted primarily with male subjects, and prevalence and severity of sleep apnea in women are underestimated. Recent clinical and basic science evidence increasingly points to different mechanisms in men and women with sleep-disordered breathing (SDB). SDB is associated with a variety of comorbidities, including cardiovascular disease, heart failure, diabetes, and atrial fibrillation. In this review, we discuss sex-dependent mechanisms of SDB in select associated conditions to sharpen our clinical understanding of these sex-dependent inherent differences.
Collapse
Affiliation(s)
- Philipp Hegner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Simon Lebek
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Lars Siegfried Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Michael Arzt
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
45
|
MacDonald CJ, Madika AL, Lajous M, Canonico M, Fournier A, Boutron-Ruault MC. Association between cardiovascular risk-factors and venous thromboembolism in a large longitudinal study of French women. Thromb J 2021; 19:58. [PMID: 34419051 PMCID: PMC8380360 DOI: 10.1186/s12959-021-00310-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/05/2021] [Indexed: 11/14/2022] Open
Abstract
Background Previous studies have shown conflicting results regarding the influence of cardiovascular risk-factors on venous thromboembolism. This study aimed to determine if these risk-factors, i.e. physical activity, smoking, hypertension, dyslipidaemia, and diabetes, were associated with the risk of venous thromboembolism, and to determine if these associations were confounded by BMI. Methods We used data from the E3N cohort study, a French prospective population-based study initiated in 1990, consisting of 98,995 women born between 1925 and 1950. From the women in the study we included those who did not have prevalent arterial disease or venous thromboembolism at baseline; thus 91,707 women were included in the study. Venous thromboembolism cases were self-reported during follow-up, and verified via specific mailings to medical practitioners or via drug reimbursements for anti-thrombotic medications. Hypertension, diabetes and dyslipidaemia were self-reported validated against drug reimbursements or specific questionnaires. Physical activity, and smoking were based on self-reports. Cox-models, adjusted for BMI and other potential risk-factors were used to determine hazard ratios for incident venous thromboembolism. Results During 1,897,960 person-years (PY), 1, 649 first incident episodes of thrombosis were identified at an incidence rate of 0.9 per 1000 PY. This included 505 cases of pulmonary embolism and 1144 cases of deep vein thrombosis with no evidence of pulmonary embolism. Hypertension, dyslipidaemia, diabetes, smoking and physical activity were not associated with the overall risk of thrombosis after adjustment for BMI. Conclusions Traditional cardiovascular risk factors were not associated with the risk of venous thromboembolism after adjustment for BMI. Hypertension, dyslipidaemia and diabetes may not be risk-factors for venous thromboembolism. Supplementary Information The online version contains supplementary material available at 10.1186/s12959-021-00310-w.
Collapse
Affiliation(s)
- C J MacDonald
- INSERM (Institut National de la Santé et de la Recherche Médicale) U1018, Center for Research in Epidemiology and Population Health (CESP), Institut Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Université Paris-Sud, Villejuif, France
| | - A L Madika
- INSERM (Institut National de la Santé et de la Recherche Médicale) U1018, Center for Research in Epidemiology and Population Health (CESP), Institut Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Université Paris-Sud, Villejuif, France.,Université de Lille, CHU Lille, EA 2694 - Santé publique : épidémiologie et qualité des soins, F-59000, Lille, France
| | - M Lajous
- Center for Research on Population Health, INSP (Instituto Nacional de Salud Pública), Cuernavaca, Mexico.,Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - M Canonico
- INSERM (Institut National de la Santé et de la Recherche Médicale) U1018, Center for Research in Epidemiology and Population Health (CESP), Institut Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Université Paris-Sud, Villejuif, France
| | - A Fournier
- INSERM (Institut National de la Santé et de la Recherche Médicale) U1018, Center for Research in Epidemiology and Population Health (CESP), Institut Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Université Paris-Sud, Villejuif, France
| | - M C Boutron-Ruault
- INSERM (Institut National de la Santé et de la Recherche Médicale) U1018, Center for Research in Epidemiology and Population Health (CESP), Institut Gustave Roussy, Villejuif, France. .,Université Paris-Saclay, Université Paris-Sud, Villejuif, France.
| |
Collapse
|
46
|
Endothelial dysfunction and thromboembolism in children, adolescents, and young adults with acute lymphoblastic leukemia. Leukemia 2021; 36:361-369. [PMID: 34389803 DOI: 10.1038/s41375-021-01383-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 07/22/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023]
Abstract
Endothelial dysfunction has not previously been investigated as a thrombogenic risk factor among patients with acute lymphoblastic leukemia (ALL), known to be at high risk of thromboembolism. We retrospectively explored the association between three circulating biomarkers of endothelial dysfunction (thrombomodulin, syndecan-1, VEGFR-1) measured in prospectively collected blood samples and risk of thromboembolism in 55 cases and 165 time-matched controls, treated according to the NOPHO ALL2008 protocol. In age-, sex-, and risk group-adjusted analysis, increasing levels of thrombomodulin and VEGFR-1 were independently associated with increased odds of developing thromboembolism (OR 1.37 per 1 ng/mL [95% CI 1.20‒1.56, P < 0.0001] and OR 1.21 per 100 pg/mL [95% CI 1.02‒1.21, P = 0.005], respectively). These associations remained significant when including only samples drawn >30 days before thromboembolic diagnosis. Thrombomodulin levels were on average 3.2 ng/mL (95% CI 2.6-8.2 ng/mL) higher in samples with measurable asparaginase activity (P < 0.0001). Among single nucleotide variants located in or neighboring coding genes for the three biomarkers, none were significantly associated with odds of thromboembolism. If results are validated in another cohort, thrombomodulin and VEGFR-1 could serve as predictive biomarkers, identifying patients in need of preemptive antithrombotic prophylaxis.
Collapse
|
47
|
Su Y, Li Q, Zheng Z, Wei X, Hou P. Identification of genes, pathways and transcription factor-miRNA-target gene networks and experimental verification in venous thromboembolism. Sci Rep 2021; 11:16352. [PMID: 34381164 PMCID: PMC8357955 DOI: 10.1038/s41598-021-95909-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 08/02/2021] [Indexed: 12/17/2022] Open
Abstract
Venous thromboembolism (VTE) is a complex, multifactorial life-threatening disease that involves vascular endothelial cell (VEC) dysfunction. However, the exact pathogenesis and underlying mechanisms of VTE are not completely clear. The aim of this study was to identify the core genes and pathways in VECs that are involved in the development and progression of unprovoked VTE (uVTE). The microarray dataset GSE118259 was downloaded from the Gene Expression Omnibus database, and 341 up-regulated and 8 down-regulated genes were identified in the VTE patients relative to the healthy controls, including CREB1, HIF1α, CBL, ILK, ESM1 and the ribosomal protein family genes. The protein-protein interaction (PPI) network and the transcription factor (TF)-miRNA-target gene network were constructed with these differentially expressed genes (DEGs), and visualized using Cytoscape software 3.6.1. Eighty-nine miRNAs were predicted as the targeting miRNAs of the DEGs, and 197 TFs were predicted as regulators of these miRNAs. In addition, 237 node genes and 4 modules were identified in the PPI network. The significantly enriched pathways included metabolic, cell adhesion, cell proliferation and cellular response to growth factor stimulus pathways. CREB1 was a differentially expressed TF in the TF-miRNA-target gene network, which regulated six miRNA-target gene pairs. The up-regulation of ESM1, HIF1α and CREB1 was confirmed at the mRNA and protein level in the plasma of uVTE patients. Taken together, ESM1, HIF1α and the CREB1-miRNA-target genes axis play potential mechanistic roles in uVTE development.
Collapse
Affiliation(s)
- Yiming Su
- Department of Vascular Surgery, LiuzhouWorker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545005, Guangxi Province, China
| | - Qiyi Li
- Department of Vascular Surgery, LiuzhouWorker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545005, Guangxi Province, China
| | - Zhiyong Zheng
- Department of Vascular Surgery, LiuzhouWorker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545005, Guangxi Province, China
| | - Xiaomin Wei
- Department of Vascular Surgery, LiuzhouWorker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545005, Guangxi Province, China
| | - Peiyong Hou
- Department of Vascular Surgery, LiuzhouWorker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, 545005, Guangxi Province, China.
| |
Collapse
|
48
|
Pan Z, Zhang Y, Li C, Yin Y, Liu R, Zheng G, Fan W, Zhang Q, Song Z, Guo Z, Rong J, Shen Y. MiR-296-5p ameliorates deep venous thrombosis by inactivating S100A4. Exp Biol Med (Maywood) 2021; 246:2259-2268. [PMID: 34192971 DOI: 10.1177/15353702211023034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Deep venous thrombosis is one of the most common venous thromboembolic diseases and has a low cure rate and a high postoperative recurrence rate. Furthermore, emerging evidence indicates that microRNAs are involved in deep venous thrombosis. miR-296-5p is an important microRNA that plays a critical role in various cellular functions, and S100A4 is closely related to vascular function. miR-296-5p is downregulated in deep venous thrombosis patients, and its predicted target S100A4 is upregulated in deep venous thrombosis patients. Therefore, it was hypothesized that miR-296-5p may play a vital role in the development of deep venous thrombosis by targeting S100A4. An Ox-LDL-stimulated HUVEC and deep venous thrombosis mouse model was employed to detect the biological functions of miR-296-5p and S100A4. Dual luciferase reporter assays and pull-down assays were used to authenticate the interaction between miR-296-5p and S100A4. ELISA and Western blotting were employed to detect the protein levels of thrombosis-related factors and the endothelial-to-mesenchymal transition (EndMT)-related factors. The miR-296-5p levels were reduced, while the S100A4 levels were enhanced in deep venous thrombosis patients, and the miR-296-5p levels were negatively correlated with the S100A4 levels in deep venous thrombosis patients. miR-296-5p suppressed S100A4 expression by targeting the 3' UTR of S100A4. MiR-296-5p knockdown accelerated ox-LDL-induced HUVEC apoptosis, oxidative stress, thrombosis-related factor expression, and EndMT, while S100A4 knockdown antagonized these effects in ox-LDL-induced HUVECs. S100A4 knockdown reversed the effect induced by miR-296-5p knockdown. Moreover, the in vivo studies revealed that miR-296-5p knockdown in deep venous thrombosis mice exacerbated deep venous thrombosis formation, whereas S100A4 knockdown had the opposite effect. These results indicate that elevated miR-296-5p inhibits deep venous thrombosis formation by inhibiting S100A4 expression. Both miR-296-5p and S100A4 may be potential diagnostic markers and therapeutic targets for deep venous thrombosis.
Collapse
Affiliation(s)
- Zhichang Pan
- Department of Vascular Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, China
| | - Yu Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Chuanyong Li
- Department of Vascular Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, China
| | - Yuan Yin
- Department of Endocrinology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, China
| | - Rui Liu
- Department of Rheumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, China
| | - Guangfeng Zheng
- Department of Vascular Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, China
| | - Weijian Fan
- Department of Vascular Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, China
| | - Qiang Zhang
- Department of Vascular Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, China
| | - Zhenyu Song
- Department of Vascular Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, China
| | - Ziyue Guo
- Department of Vascular Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, China
| | - Jianjie Rong
- Department of Vascular Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, China
| | - Yixin Shen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
49
|
Visser MJE, Venter C, Roberts TJ, Tarr G, Pretorius E. Psoriatic disease is associated with systemic inflammation, endothelial activation, and altered haemostatic function. Sci Rep 2021; 11:13043. [PMID: 34158537 PMCID: PMC8219816 DOI: 10.1038/s41598-021-90684-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/10/2021] [Indexed: 02/08/2023] Open
Abstract
Psoriasis is a chronic, immune-mediated inflammatory skin disease, affecting approximately 2% of the general population, which can be accompanied by psoriatic arthritis (PsA). The condition has been associated with an increased cardiovascular burden. Hypercoagulability is a potential underlying mechanism that may contribute to the increased risk of major cardiovascular events in psoriatic individuals. Whole blood samples were collected from 20 PsA patients and 20 healthy individuals. The concentrations of inflammatory molecules (C-reactive protein, serum amyloid A, soluble intercellular adhesion molecule-1, soluble vascular cell adhesion molecule-1, and soluble P-selectin) were determined by enzyme-linked immunosorbent assays. In addition, clotting efficiency was evaluated by thromboelastography. The fibrin network architecture was also assessed by scanning electron microscopy. Elevated levels of circulating inflammatory molecules were significantly associated with the presence of psoriatic disease. Furthermore, an increased tendency towards thrombus formation was significantly predictive of disease presence. Scanning electron microscopy revealed that fibrin clots were denser in psoriatic individuals, compared to healthy controls, with an increased fibrin fibre diameter associated with psoriatic disease. Our results add to the accumulating evidence of the systemic nature of psoriasis and the subsequent risk of cardiovascular comorbidities, potentially due to an acquired hypercoagulability. We suggest that haemostatic function should be monitored carefully in psoriatic patients that present with severe disease, due to the pre-eminent risk of developing thrombotic complications.
Collapse
Affiliation(s)
- Maria J E Visser
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 MATIELAND, Stellenbosch, 7602, South Africa
| | - Chantelle Venter
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 MATIELAND, Stellenbosch, 7602, South Africa
| | - Timothy J Roberts
- 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, Liverpool, UK.,University College London Hospital NHS Foundation Trust, 250 Euston Road, London, NW1 2PB, UK
| | - Gareth Tarr
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 MATIELAND, Stellenbosch, 7602, South Africa.,Division of Rheumatology, Institute of Orthopaedics and Rheumatology, Winelands Mediclinic Orthopaedic Hospital, Stellenbosch University, Cape Town, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 MATIELAND, Stellenbosch, 7602, South Africa.
| |
Collapse
|
50
|
Lee AC, Castaneda G, Li WT, Chen C, Shende N, Chakladar J, Taub PR, Chang EY, Ongkeko WM. COVID-19 Severity Potentially Modulated by Cardiovascular-Disease-Associated Immune Dysregulation. Viruses 2021; 13:1018. [PMID: 34071557 PMCID: PMC8228164 DOI: 10.3390/v13061018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022] Open
Abstract
Patients with underlying cardiovascular conditions are particularly vulnerable to severe COVID-19. In this project, we aimed to characterize similarities in dysregulated immune pathways between COVID-19 patients and patients with cardiomyopathy, venous thromboembolism (VTE), or coronary artery disease (CAD). We hypothesized that these similarly dysregulated pathways may be critical to how cardiovascular diseases (CVDs) exacerbate COVID-19. To evaluate immune dysregulation in different diseases, we used four separate datasets, including RNA-sequencing data from human left ventricular cardiac muscle samples of patients with dilated or ischemic cardiomyopathy and healthy controls; RNA-sequencing data of whole blood samples from patients with single or recurrent event VTE and healthy controls; RNA-sequencing data of human peripheral blood mononuclear cells (PBMCs) from patients with and without obstructive CAD; and RNA-sequencing data of platelets from COVID-19 subjects and healthy controls. We found similar immune dysregulation profiles between patients with CVDs and COVID-19 patients. Interestingly, cardiomyopathy patients display the most similar immune landscape to COVID-19 patients. Additionally, COVID-19 patients experience greater upregulation of cytokine- and inflammasome-related genes than patients with CVDs. In all, patients with CVDs have a significant overlap of cytokine- and inflammasome-related gene expression profiles with that of COVID-19 patients, possibly explaining their greater vulnerability to severe COVID-19.
Collapse
Affiliation(s)
- Abby C. Lee
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA 92093, USA; (A.C.L.); (G.C.); (W.T.L.); (C.C.); (N.S.); (J.C.)
- Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA;
| | - Grant Castaneda
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA 92093, USA; (A.C.L.); (G.C.); (W.T.L.); (C.C.); (N.S.); (J.C.)
- Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA;
| | - Wei Tse Li
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA 92093, USA; (A.C.L.); (G.C.); (W.T.L.); (C.C.); (N.S.); (J.C.)
- Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA;
| | - Chengyu Chen
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA 92093, USA; (A.C.L.); (G.C.); (W.T.L.); (C.C.); (N.S.); (J.C.)
- Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA;
| | - Neil Shende
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA 92093, USA; (A.C.L.); (G.C.); (W.T.L.); (C.C.); (N.S.); (J.C.)
- Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA;
| | - Jaideep Chakladar
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA 92093, USA; (A.C.L.); (G.C.); (W.T.L.); (C.C.); (N.S.); (J.C.)
- Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA;
| | - Pam R. Taub
- Department of Medicine, Division of Cardiovascular Medicine, University of California, San Diego, CA 92093, USA;
| | - Eric Y. Chang
- Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA;
- Department of Radiology, University of California, San Diego, CA 92093, USA
| | - Weg M. Ongkeko
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, UC San Diego School of Medicine, San Diego, CA 92093, USA; (A.C.L.); (G.C.); (W.T.L.); (C.C.); (N.S.); (J.C.)
- Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA;
| |
Collapse
|