Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.680
Filter
1.
Life Sci Alliance ; 7(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38740432

ABSTRACT

Subclinical vascular impairment can be exacerbated in individuals who experience sustained inflammation after COVID-19 infection. Our study explores the prevalence and impact of autoantibodies on vascular dysfunction in healthy COVID-19 survivors, an area that remains inadequately investigated. Focusing on autoantibodies against the atypical chemokine receptor 1 (ACKR1), COVID-19 survivors demonstrated significantly elevated anti-ACKR1 autoantibodies, correlating with systemic cytokines, circulating damaged endothelial cells, and endothelial dysfunction. An independent cohort linked these autoantibodies to increased vascular disease outcomes during a median 6.7-yr follow-up. We analyzed a single-cell transcriptome atlas of endothelial cells from diverse mouse tissues, identifying enriched Ackr1 expressions in venous regions of the brain and soleus muscle vasculatures, which holds intriguing implications for tissue-specific venous thromboembolism manifestations reported in COVID-19. Functionally, purified immunoglobulin G (IgG) extracted from patient plasma did not trigger cell apoptosis or increase barrier permeability in human vein endothelial cells. Instead, plasma IgG enhanced antibody-dependent cellular cytotoxicity mediated by patient PBMCs, a phenomenon alleviated by blocking peptide or liposome ACKR1 recombinant protein. The blocking peptide uncovered that purified IgG from COVID-19 survivors possessed potential epitopes in the N-terminal extracellular domain of ACKR1, which effectively averted antibody-dependent cellular cytotoxicity. Our findings offer insights into therapeutic development to mitigate autoantibody reactivity in blood vessels in chronic inflammation.


Subject(s)
Autoantibodies , COVID-19 , SARS-CoV-2 , Humans , Autoantibodies/immunology , COVID-19/immunology , Animals , Mice , Female , Male , SARS-CoV-2/immunology , Inflammation/immunology , Middle Aged , Endothelium, Vascular/metabolism , Endothelium, Vascular/immunology , Immunoglobulin G/immunology , Immunoglobulin G/blood , Endothelial Cells/metabolism , Endothelial Cells/immunology , Adult , Aged
2.
Viruses ; 14(2)2022 01 21.
Article in English | MEDLINE | ID: mdl-35215805

ABSTRACT

The increased plasma levels of von Willebrand factor (VWF) in patients with COVID-19 was reported in many studies, and its correlation with disease severity and mortality suggest its important role in the pathogenesis of thrombosis in COVID-19. We performed histological and immunohistochemical studies of the lungs of 29 patients who died from COVID-19. We found a significant increase in the intensity of immunohistochemical reaction for VWF in the pulmonary vascular endothelium when the disease duration was more than 10 days. In the patients who had thrombotic complications, the VWF immunostaining in the pulmonary vascular endothelium was significantly more intense than in nonsurvivors without thrombotic complications. Duration of disease and thrombotic complications were found to be independent predictors of increased VWF immunostaining in the endothelium of pulmonary vessels. We also revealed that bacterial pneumonia was associated with increased VWF staining intensity in pulmonary arterial, arteriolar, and venular endothelium, while lung ventilation was an independent predictor of increased VWF immunostaining in arterial endothelium. The results of the study demonstrated an important role of endothelial VWF in the pathogenesis of thrombus formation in COVID-19.


Subject(s)
COVID-19/complications , Lung/blood supply , Venous Thrombosis/etiology , Venous Thrombosis/pathology , von Willebrand Factor/analysis , Adult , Autopsy , COVID-19/blood , Endothelium, Vascular/immunology , Female , Humans , Immunohistochemistry/methods , Lung/pathology , Male , Middle Aged , Pneumonia, Bacterial/immunology , Pulmonary Embolism , Severity of Illness Index , Venous Thrombosis/classification
3.
Int J Mol Sci ; 23(3)2022 Jan 22.
Article in English | MEDLINE | ID: mdl-35163148

ABSTRACT

Increased soluble endoglin (sENG) has been observed in human brain arteriovenous malformations (bAVMs). In addition, the overexpression of sENG in concurrence with vascular endothelial growth factor (VEGF)-A has been shown to induce dysplastic vessel formation in mouse brains. However, the underlying mechanism of sENG-induced vascular malformations is not clear. The evidence suggests the role of sENG as a pro-inflammatory modulator, and increased microglial accumulation and inflammation have been observed in bAVMs. Therefore, we hypothesized that microglia mediate sENG-induced inflammation and endothelial cell (EC) dysfunction in bAVMs. In this study, we confirmed that the presence of sENG along with VEGF-A overexpression induced dysplastic vessel formation. Remarkably, we observed increased microglial activation around dysplastic vessels with the expression of NLRP3, an inflammasome marker. We found that sENG increased the gene expression of VEGF-A, pro-inflammatory cytokines/inflammasome mediators (TNF-α, IL-6, NLRP3, ASC, Caspase-1, and IL-1ß), and proteolytic enzyme (MMP-9) in BV2 microglia. The conditioned media from sENG-treated BV2 (BV2-sENG-CM) significantly increased levels of angiogenic factors (Notch-1 and TGFß) and pERK1/2 in ECs but it decreased the level of IL-17RD, an anti-angiogenic mediator. Finally, the BV2-sENG-CM significantly increased EC migration and tube formation. Together, our study demonstrates that sENG provokes microglia to express angiogenic/inflammatory molecules which may be involved in EC dysfunction. Our study corroborates the contribution of microglia to the pathology of sENG-associated vascular malformations.


Subject(s)
Endoglin/administration & dosage , Endothelium, Vascular/pathology , Inflammation/pathology , Microglia/pathology , Neovascularization, Pathologic/pathology , Vascular Diseases/pathology , Vascular Endothelial Growth Factor A/administration & dosage , Animals , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Inflammation/chemically induced , Inflammation/immunology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Vascular Diseases/chemically induced , Vascular Diseases/immunology , Vascular Diseases/metabolism , Vascular Endothelial Growth Factor A/adverse effects
4.
Lab Chip ; 22(6): 1171-1186, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35142777

ABSTRACT

Coronavirus disease 2019 (COVID-19) was primarily identified as a novel disease causing acute respiratory syndrome. However, as the pandemic progressed various cases of secondary organ infection and damage by severe respiratory syndrome coronavirus 2 (SARS-CoV-2) have been reported, including a breakdown of the vascular barrier. As SARS-CoV-2 gains access to blood circulation through the lungs, the virus is first encountered by the layer of endothelial cells and immune cells that participate in host defense. Here, we developed an approach to study SARS-CoV-2 infection using vasculature-on-a-chip. We first modeled the interaction of virus alone with the endothelialized vasculature-on-a-chip, followed by the studies of the interaction of the virus exposed-endothelial cells with peripheral blood mononuclear cells (PBMCs). In an endothelial model grown on a permeable microfluidic bioscaffold under flow conditions, both human coronavirus (HCoV)-NL63 and SARS-CoV-2 presence diminished endothelial barrier function by disrupting VE-cadherin junctions and elevating the level of pro-inflammatory cytokines such as interleukin (IL)-6, IL-8, and angiopoietin-2. Inflammatory cytokine markers were markedly more elevated upon SARS-CoV-2 infection compared to HCoV-NL63 infection. Introduction of PBMCs with monocytes into the vasculature-on-a-chip upon SARS-CoV-2 infection further exacerbated cytokine-induced endothelial dysfunction, demonstrating the compounding effects of inter-cellular crosstalk between endothelial cells and monocytes in facilitating the hyperinflammatory state. Considering the harmful effects of SARS-CoV-2 on endothelial cells, even without active virus proliferation inside the cells, a potential therapeutic approach is critical. We identified angiopoietin-1 derived peptide, QHREDGS, as a potential therapeutic capable of profoundly attenuating the inflammatory state of the cells consistent with the levels in non-infected controls, thereby improving the barrier function and endothelial cell survival against SARS-CoV-2 infection in the presence of PBMC.


Subject(s)
Angiopoietin-1 , COVID-19 Drug Treatment , COVID-19 , Endothelium, Vascular , Inflammation , SARS-CoV-2 , COVID-19/virology , Endothelial Cells/immunology , Endothelial Cells/virology , Endothelium, Vascular/immunology , Endothelium, Vascular/virology , Humans , Immunity, Innate , Inflammation/drug therapy , Inflammation/virology , Lab-On-A-Chip Devices , Leukocytes, Mononuclear
5.
Biochem Pharmacol ; 197: 114909, 2022 03.
Article in English | MEDLINE | ID: mdl-35021044

ABSTRACT

Vascular endothelial cells are major participants in and regulators of immune responses and inflammation. Vascular endotheliitis is regarded as a host immune-inflammatory response of the endothelium forming the inner surface of blood vessels in association with a direct consequence of infectious pathogen invasion. Vascular endotheliitis and consequent endothelial dysfunction can be a principle determinant of microvascular failure, which would favor impaired perfusion, tissue hypoxia, and subsequent organ failure. Emerging evidence suggests the role of vascular endotheliitis in the pathogenesis of coronavirus disease 2019 (COVID-19) and its related complications. Thus, once initiated, vascular endotheliitis and resultant cytokine storm cause systemic hyperinflammation and a thrombotic phenomenon in COVID-19, leading to acute respiratory distress syndrome and widespread organ damage. Vascular endotheliitis also appears to be a contributory factor to vasculopathy and coagulopathy in sepsis that is defined as life-threatening organ dysfunction due to a dysregulated response of the host to infection. Therefore, protecting endothelial cells and reversing vascular endotheliitis may be a leading therapeutic goal for these diseases associated with vascular endotheliitis. In this review, we outline the etiological and pathogenic importance of vascular endotheliitis in infection-related inflammatory diseases, including COVID-19, and possible mechanisms leading to vascular endotheliitis. We also discuss pharmacological agents which may be now considered as potential endotheliitis-based treatment modalities for those diseases.


Subject(s)
COVID-19/pathology , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Vascular Diseases/pathology , COVID-19/complications , COVID-19/immunology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , Sepsis/drug therapy , Sepsis/etiology , Sepsis/immunology , Sepsis/pathology , Vascular Diseases/drug therapy , Vascular Diseases/etiology , Vascular Diseases/immunology , COVID-19 Drug Treatment
6.
J Mol Cell Cardiol ; 164: 69-82, 2022 03.
Article in English | MEDLINE | ID: mdl-34838588

ABSTRACT

The global propagation of SARS-CoV-2 leads to an unprecedented public health emergency. Despite that the lungs are the primary organ targeted by COVID-19, systemic endothelial inflammation and dysfunction is observed particularly in patients with severe COVID-19, manifested by elevated endothelial injury markers, endotheliitis, and coagulopathy. Here, we review the clinical characteristics of COVID-19 associated endothelial dysfunction; and the likely pathological mechanisms underlying the disease including direct cell entry or indirect immune overreactions after SARS-CoV-2 infection. In addition, we discuss potential biomarkers that might indicate the disease severity, particularly related to the abnormal development of thrombosis that is a fatal vascular complication of severe COVID-19. Furthermore, we summarize clinical trials targeting the direct and indirect pathological pathways after SARS-CoV-2 infection to prevent or inhibit the virus induced endothelial disorders.


Subject(s)
COVID-19/pathology , Endothelium, Vascular/pathology , SARS-CoV-2 , Adolescent , Adult , Aged , Angiotensin-Converting Enzyme 2/physiology , Animals , COVID-19/blood , COVID-19/complications , COVID-19/physiopathology , COVID-19/therapy , Clinical Trials as Topic , Endothelial Cells/pathology , Endothelial Cells/virology , Endothelium, Vascular/immunology , Endothelium, Vascular/physiopathology , HMGB1 Protein/physiology , Humans , Macaca mulatta , Mice , Neuropilin-1/physiology , Oxidative Stress , Reactive Oxygen Species , Receptors, Virus/physiology , Scavenger Receptors, Class B/physiology , Severity of Illness Index , Signal Transduction , Systemic Inflammatory Response Syndrome/pathology , Systemic Inflammatory Response Syndrome/physiopathology , Thrombophilia/etiology , Thrombophilia/physiopathology , Vascular Endothelial Growth Factor A/physiology , Vasculitis/etiology , Vasculitis/immunology , Vasculitis/physiopathology , Young Adult
7.
Cardiovasc Res ; 118(1): 254-266, 2022 01 07.
Article in English | MEDLINE | ID: mdl-33483748

ABSTRACT

AIMS: Rheumatoid arthritis (RA) is a chronic inflammatory disease affecting joints and blood vessels. Despite low levels of low-density lipoprotein cholesterol (LDL-C), RA patients exhibit endothelial dysfunction and are at increased risk of death from cardiovascular complications, but the molecular mechanism of action is unknown. We aimed in the present study to identify the molecular mechanism of endothelial dysfunction in a mouse model of RA and in patients with RA. METHODS AND RESULTS: Endothelium-dependent relaxations to acetylcholine were reduced in aortae of two tumour necrosis factor alpha (TNFα) transgenic mouse lines with either mild (Tg3647) or severe (Tg197) forms of RA in a time- and severity-dependent fashion as assessed by organ chamber myograph. In Tg197, TNFα plasma levels were associated with severe endothelial dysfunction. LOX-1 receptor was markedly up-regulated leading to increased vascular oxLDL uptake and NFκB-mediated enhanced Arg2 expression via direct binding to its promoter resulting in reduced NO bioavailability and vascular cGMP levels as shown by ELISA and chromatin immunoprecipitation. Anti-TNFα treatment with infliximab normalized endothelial function together with LOX-1 and Arg2 serum levels in mice. In RA patients, soluble LOX-1 serum levels were also markedly increased and closely related to serum levels of C-reactive protein. Similarly, ARG2 serum levels were increased. Similarly, anti-TNFα treatment restored LOX-1 and ARG2 serum levels in RA patients. CONCLUSIONS: Increased TNFα levels not only contribute to RA, but also to endothelial dysfunction by increasing vascular oxLDL content and activation of the LOX-1/NFκB/Arg2 pathway leading to reduced NO bioavailability and decreased cGMP levels. Anti-TNFα treatment improved both articular symptoms and endothelial function by reducing LOX-1, vascular oxLDL, and Arg2 levels.


Subject(s)
Aorta, Thoracic/drug effects , Arginase/metabolism , Arthritis, Rheumatoid/drug therapy , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Scavenger Receptors, Class E/metabolism , Tumor Necrosis Factor Inhibitors/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Vasodilation/drug effects , Adult , Animals , Animals, Genetically Modified , Aorta, Thoracic/enzymology , Aorta, Thoracic/immunology , Aorta, Thoracic/physiopathology , Arginase/genetics , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/physiopathology , Case-Control Studies , Disease Models, Animal , Endothelial Cells/enzymology , Endothelial Cells/immunology , Endothelium, Vascular/enzymology , Endothelium, Vascular/immunology , Endothelium, Vascular/physiopathology , Female , Humans , Lipoproteins, LDL/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred CBA , Middle Aged , NF-kappa B/metabolism , Scavenger Receptors, Class E/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/genetics
8.
Hepatol Commun ; 6(2): 255-269, 2022 02.
Article in English | MEDLINE | ID: mdl-34658172

ABSTRACT

Liver injury, characterized predominantly by elevated aspartate aminotransferase and alanine aminotransferase, is a common feature of coronavirus disease 2019 (COVID-19) symptoms caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2). Additionally, SARS-CoV-2 infection is associated with acute-on-chronic liver failure in patients with cirrhosis and has a notably elevated mortality in patients with alcohol-related liver disease compared to other etiologies. Direct viral infection of the liver with SARS-CoV-2 remains controversial, and alternative pathophysiologic explanations for its hepatic effects are an area of active investigation. In this review, we discuss the effects of SARS-CoV-2 and the inflammatory environment it creates on endothelial cells and platelets more generally and then with a hepatic focus. In doing this, we present vascular inflammation and thrombosis as a potential mechanism of liver injury and liver-related complications in COVID-19.


Subject(s)
Blood Platelet Disorders/virology , COVID-19/physiopathology , Endothelium, Vascular/virology , Inflammation/virology , Liver Diseases/virology , Thrombosis/virology , Blood Platelet Disorders/immunology , Blood Platelet Disorders/physiopathology , COVID-19/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/physiopathology , Humans , Inflammation/immunology , Inflammation/physiopathology , Liver Diseases/immunology , Liver Diseases/physiopathology , Thrombosis/immunology , Thrombosis/physiopathology
9.
Inflammation ; 45(1): 116-128, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34494203

ABSTRACT

Angioedema is characterized by swelling of the skin or mucous membranes. Overproduction of the vasodilator bradykinin (BK) is an important contributor to the disease pathology, which causes rapid increase in vascular permeability. BK formation on endothelial cells results from high molecular weight kininogen (HK) interacting with gC1qR, the receptor for the globular heads of C1q, the first component of the classical pathway of complement. Endothelial cells are sensitive to blood-flow-induced shear stress and it has been shown that shear stress can modulate gC1qR expression. This study aimed to determine the following: (1) how BK or angioedema patients' (HAE) plasma affected endothelial cell permeability and gC1qR expression under shear stress, and (2) if monoclonal antibody (mAb) 74.5.2, which recognizes the HK binding site on gC1qR, had an inhibitory effect in HK binding to endothelial cells. Human dermal microvascular endothelial cells (HDMECs) grown on Transwell inserts were exposed to shear stress in the presence of HAE patients' plasma. Endothelial cell permeability was measured using FITC-conjugated bovine serum albumin. gC1qR expression and HK binding to endothelial cell surface was measured using solid-phase ELISA. Cell morphology was quantified using immunofluorescence microscopy. The results demonstrated that BK at 1 µg/mL, but not HAE patients' plasma and/or shear stress, caused significant increases in HDMEC permeability. The mAb 74.5.2 could effectively inhibit HK binding to recombinant gC1qR, and reduce HAE patients' plasma-induced HDMEC permeability change. These results suggested that monoclonal antibody to gC1qR, i.e., 74.5.2, could be potentially used as an effective therapeutic reagent to prevent angioedema.


Subject(s)
Angioedema/drug therapy , Antibodies, Monoclonal/pharmacology , Bradykinin/metabolism , Capillary Permeability/drug effects , Cardiovascular Agents/pharmacology , Carrier Proteins/immunology , Endothelial Cells/drug effects , Mitochondrial Proteins/immunology , Angioedema/immunology , Angioedema/metabolism , Angioedema/physiopathology , Antibodies, Monoclonal/therapeutic use , Biomarkers/metabolism , Capillary Permeability/immunology , Cardiovascular Agents/therapeutic use , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Humans , Permeability/drug effects , Shear Strength/drug effects
10.
Eur J Immunol ; 52(2): 204-221, 2022 02.
Article in English | MEDLINE | ID: mdl-34708880

ABSTRACT

Multiple embryonic precursors give rise to leukocytes in adults while the lineage-based functional impacts are underappreciated. Mesodermal precursors expressing PDGFRα appear transiently during E7.5-8.5 descend to a subset of Lin- Sca1+ Kit+ hematopoietic progenitors found in adult BM. By analyzing a PDGFRα-lineage tracing mouse line, we here report that PDGFRα-lineage BM F4/80+ SSClo monocytes/macrophages are solely Ly6C+ LFA-1hi Mac-1hi monocytes enriched on the abluminal sinusoidal endothelium while Ly6C- LFA-1lo Mac-1lo macrophages are mostly from non-PDGFRα-lineage in vivo. Monocytes with stronger integrin profiles outcompete macrophages for adhesion on an endothelial monolayer or surfaces coated with ICAM-1-Fc or VCAM-1-Fc. Egress of PDGFRα-lineage-rich monocytes and subsequent differentiation to peripheral macrophages spatially segregates them from non-PDGFRα-lineage BM-resident macrophages and allows functional specialization since macrophages derived from these egressing monocytes differ in morphology, phenotype, and functionality from BM-resident macrophages in culture. Extravasation preference for blood PDGFRα-lineage monocytes varies by tissues and governs the local lineage composition of macrophages. More PDGFRα-lineage classical monocytes infiltrated into skin and colon but not into peritoneum. Accordingly, transcriptomic analytics indicated augmented inflammatory cascades in dermatitis skin of BM-chimeric mice harbouring only PDGFRα-lineage leukocytes. Thus, the PDGFRα-lineage origin biasedly generates monocytes predestined for BM exit to support peripheral immunity following extravasation and macrophage differentiation.


Subject(s)
Cell Lineage/immunology , Cell Movement/immunology , Endothelium, Vascular/immunology , Macrophages/immunology , Monocytes/immunology , Receptor, Platelet-Derived Growth Factor alpha/immunology , Animals , Cell Lineage/genetics , Cell Movement/genetics , Mice , Mice, Transgenic , Receptor, Platelet-Derived Growth Factor alpha/genetics
11.
Front Immunol ; 12: 770436, 2021.
Article in English | MEDLINE | ID: mdl-34970260

ABSTRACT

The article presents the function of platelets in inflammation as well as in bacterial and viral infections, which are the result of their reaction with the endovascular environment, including cells of damaged vascular endothelium and cells of the immune system. This role of platelets is conditioned by biologically active substances present in their granules and in their specific structures - EV (extracellular vesicles).


Subject(s)
Bacterial Infections/immunology , Blood Platelets/immunology , Extracellular Vesicles/immunology , Immune System/immunology , Inflammation/immunology , Virus Diseases/immunology , Bacterial Infections/microbiology , Blood Platelets/metabolism , Cytokines/immunology , Cytokines/metabolism , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Humans , Immune System/microbiology , Immune System/virology , Inflammation/metabolism , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Models, Immunological , Signal Transduction/immunology , Virus Diseases/virology
12.
Scand J Immunol ; 94(5): e13097, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34940978

ABSTRACT

COVID-19 is a global pandemic with a daily increasing number of affected individuals. Thrombosis is a severe complication of COVID-19 that leads to a worse clinical course with higher rates of mortality. Multiple lines of evidence suggest that hyperinflammation plays a crucial role in disease progression. This review compiles clinical data of COVID-19 patients who developed thrombotic complications to investigate the possible role of hyperinflammation in inducing hypercoagulation. A systematic literature search was performed using PubMed, Embase, Medline and Scopus to identify relevant clinical studies that investigated thrombotic manifestations and reported inflammatory and coagulation biomarkers in COVID-19 patients. Only 54 studies met our inclusion criteria, the majority of which demonstrated significantly elevated inflammatory markers. In the cohort studies with control, D-dimer was significantly higher in COVID-19 patients with thrombosis as compared to the control. Pulmonary embolism, deep vein thrombosis and strokes were frequently reported which could be attributed to the hyperinflammatory response associated with COVID-19 and/or to the direct viral activation of platelets and endothelial cells, two mechanisms that are discussed in this review. It is recommended that all admitted COVID-19 patients should be assessed for hypercoagulation. Furthermore, several studies have suggested that anticoagulation may be beneficial, especially in hospitalized non-ICU patients. Although vaccines against SARS-CoV-2 have been approved and distributed in several countries, research should continue in the field of prevention and treatment of COVID-19 and its severe complications including thrombosis due to the emergence of new variants against which the efficacy of the vaccines is not yet clear.


Subject(s)
Arteries/pathology , Blood Platelets/immunology , COVID-19/immunology , Endothelium, Vascular/immunology , Inflammation/immunology , SARS-CoV-2/physiology , Venous Thrombosis/immunology , Animals , Anticoagulants/therapeutic use , Blood Platelets/virology , COVID-19/complications , Endothelium, Vascular/virology , Humans , Inflammation/complications , Phenotype , Thrombosis , Venous Thrombosis/etiology , Venous Thrombosis/prevention & control
13.
Signal Transduct Target Ther ; 6(1): 418, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34893580

ABSTRACT

The systemic processes involved in the manifestation of life-threatening COVID-19 and in disease recovery are still incompletely understood, despite investigations focusing on the dysregulation of immune responses after SARS-CoV-2 infection. To define hallmarks of severe COVID-19 in acute disease (n = 58) and in disease recovery in convalescent patients (n = 28) from Hannover Medical School, we used flow cytometry and proteomics data with unsupervised clustering analyses. In our observational study, we combined analyses of immune cells and cytokine/chemokine networks with endothelial activation and injury. ICU patients displayed an altered immune signature with prolonged lymphopenia but the expansion of granulocytes and plasmablasts along with activated and terminally differentiated T and NK cells and high levels of SARS-CoV-2-specific antibodies. The core signature of seven plasma proteins revealed a highly inflammatory microenvironment in addition to endothelial injury in severe COVID-19. Changes within this signature were associated with either disease progression or recovery. In summary, our data suggest that besides a strong inflammatory response, severe COVID-19 is driven by endothelial activation and barrier disruption, whereby recovery depends on the regeneration of the endothelial integrity.


Subject(s)
Antibodies, Viral/blood , Blood Proteins/metabolism , COVID-19/diagnosis , Cytokine Release Syndrome/diagnosis , Endothelium, Vascular/virology , Lymphopenia/diagnosis , SARS-CoV-2/pathogenicity , Biomarkers/blood , C-Reactive Protein/metabolism , COVID-19/immunology , COVID-19/mortality , COVID-19/virology , Chemokine CXCL10/blood , Chemokine CXCL9/blood , Cluster Analysis , Convalescence , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/mortality , Cytokine Release Syndrome/virology , Disease Progression , Endothelium, Vascular/immunology , Granulocytes/immunology , Granulocytes/virology , Hematopoietic Cell Growth Factors/blood , Hepatocyte Growth Factor/blood , Humans , Intensive Care Units , Interleukin-12 Subunit p40/blood , Interleukin-6/blood , Interleukin-8/blood , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Lectins, C-Type/blood , Lymphopenia/immunology , Lymphopenia/mortality , Lymphopenia/virology , Plasma Cells/immunology , Plasma Cells/virology , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/virology
14.
Sci Rep ; 11(1): 21439, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34728714

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of death in patients with chronic kidney disease (CKD). Endothelial cell (EC) dysfunction is a key CKD-specific risk factor; however, the mechanisms by which uremia harms the endothelium are still unclear. We report a role for excessive neutrophil extracellular trap (NET) formation induced by uremic serum on EC injury. Level of plasma nucleosome and myeloperoxidase-DNA, established in vivo markers of NETs, as well as intracellular adhesion molecule (ICAM)-1 were measured in hemodialysis (HD) patients and healthy volunteers (HV) and their prognostic role evaluated. For in vitro studies, HV-derived neutrophils and differentiated HL-60 cells by retinoic acid were used to determine the effect of uremic serum-induced NETs on human umbilical vein EC (HUVEC). The level of in vivo NETs was significantly higher in incident HD patients compared to HV, and these markers were strongly associated with ICAM-1. Specifically, nucleosome and ICAM-1 levels were independent predictors of a composite endpoint, all-cause mortality, or vascular access failure. In vitro, HD-derived uremic serum significantly increased NET formation both in dHL-60 and isolated neutrophils compared to control serum, and these NETs decreased EC viability and induced their apoptosis. In addition, the level of ICAM-1, E-selectin and von Willebrand factor in HUVEC supernatant was significantly increased by uremic serum-induced NETs compared to control serum-induced NETs. Dysregulated neutrophil activities in the uremic milieu may play a key role in vascular inflammatory responses. The high mortality and CVD rates in ESRD may be explained in part by excessive NET formation leading to EC damage and dysfunction.


Subject(s)
Endothelium, Vascular/pathology , Extracellular Traps/immunology , Intercellular Adhesion Molecule-1/metabolism , Renal Dialysis/adverse effects , Renal Insufficiency, Chronic/therapy , Uremia/pathology , Vascular Diseases/pathology , Aged , Case-Control Studies , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Extracellular Traps/metabolism , Female , HL-60 Cells , Humans , Male , Renal Insufficiency, Chronic/pathology , Uremia/blood , Uremia/etiology , Vascular Diseases/etiology
15.
Isr Med Assoc J ; 23(10): 657-661, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34672449

ABSTRACT

BACKGROUND: Anti-endothelial cell antibodies (AECA) are a known biomarker of endothelial dysfunction and damage in clinical practice, especially in autoimmune disease. OBJECTIVES: To determine the relation between natural AECA levels and prognosis related to coronary artery disease. METHODS: Candidates for coronary angiography were prospectively enrolled. AECA levels were determined by ELISA assay. Mortality was evaluated after more than 5 years follow-up. RESULTS: Of a total 857 patients, 445 had high AECA levels (group 1) and 412 had low levels (< 1 OD unit, group 2). Both groups did not differ in age, sex, or presence of diabetes. The median follow up was 2293 days (76 months). Patients with high AECA levels were more likely to have normal coronary arteries on angiography (21.6% vs. 16.9%, P = 0.047) and less likely to have calcified lesions (19.0% vs. 26.6%, P = 0.028) and lower prevalence of abnormal renal functions (71.1 mg/dl vs. 66.5 mg/dl, P = 0.033). Patients with higher AECA levels had lower mortality levels (20.1% vs. 27.6%, P = 0.006). A logistic regression model demonstrated independent association between lower AECA levels and the presence of coronary atherosclerosis based on angiogram. CONCLUSIONS: After a median of more than 6 years, higher natural AECA levels were associated with less coronary artery disease and lower mortality rates in patients undergoing coronary angiography.


Subject(s)
Autoantibodies/blood , Coronary Angiography , Coronary Artery Disease , Endothelium, Vascular , Aged , Biomarkers/blood , Coronary Angiography/methods , Coronary Angiography/statistics & numerical data , Coronary Artery Disease/diagnosis , Coronary Artery Disease/immunology , Coronary Artery Disease/mortality , Endothelium, Vascular/immunology , Endothelium, Vascular/physiopathology , Female , Follow-Up Studies , Humans , Immunologic Tests/methods , Israel/epidemiology , Male , Prognosis , Protective Factors , Reproducibility of Results , Risk Assessment/methods
16.
Molecules ; 26(19)2021 Sep 26.
Article in English | MEDLINE | ID: mdl-34641380

ABSTRACT

Endothelial dysfunction and inflammation are recognised factors in the development of atherosclerosis. Evidence suggests that intake of industrial trans fatty acids (TFAs) promotes endothelial dysfunction, while ruminant TFAs may have the opposite effect. The aim of this study was to compare the effects of elaidic acid (EA (18:1n-9t); an industrially produced TFA) and trans vaccenic acid (TVA (18:1n-7t); a natural TFA found in ruminant milk and meat) on inflammatory responses of endothelial cells (ECs). ECs (EA.hy926 cells) were cultured under standard conditions and exposed to TFAs (1 to 50 µM) for 48 h. Then, the cells were cultured for a further 6 or 24 h with tumour necrosis factor alpha (TNF-α, 1 ng/mL) as an inflammatory stimulant. ECs remained viable after treatments. TFAs were incorporated into ECs in a dose-dependent manner. Preincubation with EA (50 µM) increased production of MCP-1, RANTES, and IL-8 in response to TNF-α, while preincubation with TVA (1 µM) decreased production of ICAM-1 and RANTES in response to TNF-α. Preincubation with EA (50 µM) upregulated toll-like receptor 4 and cyclooxygenase 2 gene expression in response to TNF-α. In contrast, preincubation with TVA (1 µM) downregulated TNF-α induced nuclear factor kappa B subunit 1 gene expression. Preincubation of ECs with EA (50 µM) increased THP-1 monocyte adhesion. In contrast, preincubation of ECs with TVA (1 µM) reduced THP-1 monocyte adhesion, while preincubation of ECs with TVA (50 µM) decreased the level of surface expression of ICAM-1 seen following TNF-α stimulation. The results suggest that TVA has some anti-inflammatory properties, while EA enhances the response to an inflammatory stimulus. These findings suggest differential effects induced by the TFAs tested, fitting with the idea that industrial TFAs and ruminant TFAs can have different and perhaps opposing biological actions in an inflammatory context.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Carbon Radioisotopes/analysis , Endothelium, Vascular/immunology , Inflammation/immunology , Oleic Acids/pharmacology , Ruminants/metabolism , Trans Fatty Acids/pharmacology , Animals , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Tumor Necrosis Factor-alpha/metabolism
17.
Trends Endocrinol Metab ; 32(11): 875-889, 2021 11.
Article in English | MEDLINE | ID: mdl-34593305

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a pandemic of respiratory and cardiovascular diseases, known as coronavirus disease 2019 (COVID-19). SARS-CoV-2 encodes the structural proteins spike (S), envelope (E), membrane (M), and nucleocapsid (N). The receptor-binding domain on the surface subunit S1 is responsible for attachment of the virus to angiotensin (Ang)-converting enzyme 2 (ACE2), which is highly expressed in host cells. The cytokine storm observed in patients with COVID-19 contributes to the endothelial vascular dysfunction, which can lead to acute respiratory distress syndrome, multiorgan failure, alteration in iron homeostasis, and death. Growth and differentiation factor 15 (GDF15), which belongs to the transforming growth factor-ß (TGF-ß) superfamily of proteins, has a pivotal role in the development and progression of diseases because of its role as a metabolic regulator. In COVID-19, GDF15 activity increases in response to tissue damage. GDF15 appears to be a strong predictor of poor outcomes in patients critically ill with COVID-19 and acts as an 'inflammation-induced central mediator of tissue tolerance' via its metabolic properties. In this review, we examine the potential properties of GDF15 as an emerging modulator of immunity in COVID-19 in association with iron metabolism. The virus life cycle in host cell provides potential targets for drug therapy.


Subject(s)
COVID-19/immunology , Cytokine Release Syndrome/immunology , Endothelium, Vascular/immunology , Growth Differentiation Factor 15/immunology , Iron/metabolism , Apoptosis/immunology , COVID-19/metabolism , Cytokine Release Syndrome/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Glial Cell Line-Derived Neurotrophic Factor Receptors/immunology , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Growth Differentiation Factor 15/metabolism , Humans , Immunologic Factors/therapeutic use , Oxidative Stress/immunology , Prognosis , Pyroptosis/immunology , SARS-CoV-2 , COVID-19 Drug Treatment
18.
Eur J Pharmacol ; 910: 174490, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34492283

ABSTRACT

Stent-induced vascular injury is manifested by removal of the endothelium and phenotypic changes in the underlying medial smooth muscle cells layer. This results in pathological vascular remodelling primarily contributed to smooth muscle cell proliferation and leads to vessel re-narrowing; neointimal hyperplasia. Current drug-eluting stents release non-selective anti-proliferative drugs such as paclitaxel from the stent surface that not only inhibit growth of smooth muscle cells but also delay endothelial healing, potentially leading to stent thrombosis. This highlights the need for novel bioactive stent coating candidates with the ability to target key events in the pathogenesis of in-stent restenosis. Citric acid, a molecule with anti-coagulant properties, was investigated against L-ascorbic acid, an antioxidant molecule reported to preferentially promote endothelial growth, and paclitaxel, a typically used anti-proliferative stent coating. Citric acid was found to exhibit growth supporting properties on endothelial cells across a range of concentrations that were significantly better than the model stent coating drug paclitaxel and better than the ascorbic acid which inhibited endothelial proliferation at concentrations ≥100 µg/ml. It was demonstrated that a citric acid-paclitaxel combination treatment significantly improves cell viability in comparison to paclitaxel only treated cells, with endothelial cells exhibiting greater cell recovery over smooth muscle cells. Furthermore, cell treatment with citric acid was found to reduce inflammation in a lipopolysaccharide (LPS)-induced in vitro inflammation model by significantly reducing interleukin 6 expression. Thus, this study demonstrates that citric acid is a promising candidate for use as a coating in stents and other endovascular devices.


Subject(s)
Citric Acid/administration & dosage , Drug-Eluting Stents/adverse effects , Thrombosis/prevention & control , Animals , Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Humans , Mice , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/pathology , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Thrombosis/chemically induced , Thrombosis/immunology , Thrombosis/pathology , Vascular Remodeling/drug effects
19.
Int J Mol Sci ; 22(18)2021 Sep 16.
Article in English | MEDLINE | ID: mdl-34576169

ABSTRACT

Autotaxin (ATX; ENPP2) is a secreted lysophospholipase D catalyzing the extracellular production of lysophosphatidic acid (LPA), a pleiotropic signaling phospholipid. Genetic and pharmacologic studies have previously established a pathologic role for ATX and LPA signaling in pulmonary injury, inflammation, and fibrosis. Here, increased ENPP2 mRNA levels were detected in immune cells from nasopharyngeal swab samples of COVID-19 patients, and increased ATX serum levels were found in severe COVID-19 patients. ATX serum levels correlated with the corresponding increased serum levels of IL-6 and endothelial damage biomarkers, suggesting an interplay of the ATX/LPA axis with hyperinflammation and the associated vascular dysfunction in COVID-19. Accordingly, dexamethasone (Dex) treatment of mechanically ventilated patients reduced ATX levels, as shown in two independent cohorts, indicating that the therapeutic benefits of Dex include the suppression of ATX. Moreover, large scale analysis of multiple single cell RNA sequencing datasets revealed the expression landscape of ENPP2 in COVID-19 and further suggested a role for ATX in the homeostasis of dendritic cells, which exhibit both numerical and functional deficits in COVID-19. Therefore, ATX has likely a multifunctional role in COVID-19 pathogenesis, suggesting that its pharmacological targeting might represent an additional therapeutic option, both during and after hospitalization.


Subject(s)
COVID-19/diagnosis , Dendritic Cells/immunology , Phosphodiesterase Inhibitors/therapeutic use , Phosphoric Diester Hydrolases/blood , SARS-CoV-2/immunology , Adult , Aged , Aged, 80 and over , Biomarkers/blood , COVID-19/blood , COVID-19/immunology , COVID-19/therapy , Cohort Studies , Datasets as Topic , Dendritic Cells/drug effects , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Female , Humans , Interleukin-6/blood , Interleukin-6/metabolism , Male , Middle Aged , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , RNA-Seq , Respiration, Artificial , SARS-CoV-2/isolation & purification , Severity of Illness Index , Signal Transduction/drug effects , Signal Transduction/immunology , Single-Cell Analysis
20.
J Dermatol Sci ; 104(1): 63-73, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34556381

ABSTRACT

BACKGROUND: CXCL4, a chemokine with anti-angiogenic property, is involved in systemic sclerosis (SSc) related pulmonary arterial hypertension (PAH). OBJECTIVE: To investigated the contribution of CXCL4 to SSc development by focusing on the correlation of circulatory CXCL4 levels with their peripheral vasculopathy, and the effect of CXCL4 on endothelial cell dysfunction and the potential signaling. METHODS: We measured the plasma CXCL4 levels in 58 patients with SSc, 10 patients with the very early diagnosis of SSc (VEDOSS), and 80 healthy controls (HCs). Then, CXCL4 concentrations were correlated with clinical features, especially the peripheral vasculopathy. These observations were further validated in an additional cohort. Moreover, we studied the anti-angiogenic effects of CXCL4 and the underlying downstream signaling in human umbilical vein endothelial cells (HUVECs) in vitro. RESULTS: Circulating CXCL4 levels were 103.62 % higher in patients with SSc and 201.51 % higher in patients with VEDOSS than matched HCs, which were confirmed in two independent cohorts. CXCL4 levels were associated with digital ulcers (DU) and nailfold videocapillaroscopy (NVC) abnormalities in SSc. The proliferation, migration, and tube formation of HUVECs were inhibited by CXCL4 or SSc derived plasma, which reversed by CXCL4 neutralizing antibody, but failed by CXCR3 inhibitor. CXCL4 downregulated the transcription factor Friend leukaemia integration factor-1 (Fli-1) via c-Abl signaling. Furthermore, CXCL4 blocked the transforming growth factor (TGF) -ß or platelet-derived growth factor (PDGF) induced cell proliferation of HUVECs. CONCLUSIONS: CXCL4 may contribute to peripheral vasculopathy in SSc by downregulating Fli-1 via c-Abl signaling in endothelial cells and interfering angiogenesis.


Subject(s)
Endothelium, Vascular/pathology , Foot Ulcer/immunology , Platelet Factor 4/metabolism , Raynaud Disease/immunology , Scleroderma, Systemic/complications , Adult , Aged , Case-Control Studies , Cell Movement , Cell Proliferation , Early Diagnosis , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Female , Foot Ulcer/blood , Foot Ulcer/diagnosis , Foot Ulcer/pathology , Healthy Volunteers , Human Umbilical Vein Endothelial Cells , Humans , Male , Microscopic Angioscopy , Middle Aged , Platelet Factor 4/blood , Proto-Oncogene Protein c-fli-1/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Raynaud Disease/blood , Raynaud Disease/diagnosis , Raynaud Disease/pathology , Scleroderma, Systemic/blood , Scleroderma, Systemic/immunology , Scleroderma, Systemic/pathology , Signal Transduction/immunology , Skin/blood supply , Skin/diagnostic imaging , Skin/immunology , Skin/pathology , THP-1 Cells , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...