1
|
Kuruca N, Atilla A, Kaya MT, Gokmen S, Nursal AF, Kilic O, Kuruoglu T, Temocin F, Guvenc T, Yigit S, Guvenc D. Effect of VDR and TLR2 gene variants on the clinical course of patients with COVID-19 disease. J Investig Med 2024; 72:876-882. [PMID: 39075671 DOI: 10.1177/10815589241270418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, which has caused a major global health crisis, primarily targets the upper and lower respiratory tract. But infected individuals may experience different clinical symptoms, ranging from asymptomatic to critical. The vitamin D receptor (VDR) and Toll-like receptor 2 (TLR2) polymorphisms play a role in the immune response. This study aimed to evaluate the effect of VDR Bsml (rs1544410) and TLR2 23bp indel variants on the clinical status of Turkish patients with COVID-19 disease. A total of 312 people, including 106 intensive care unit (ICU) patients, 103 symptomatic hospitalized patients, and 103 healthy controls, were included in the study. The VDR BsmI and TLR2 23bp indel were genotyped using polymerase chain reaction and/or restriction fragment length fraction methods. The VDR BsmI b/b genotype and b allele were higher in symptomatic patients compared to the healthy control group (p = 0.035). The VDR BsmI B/B and B/b genotype distribution did not differ between ICU patients and both symptomatic patients and controls (p > 0.05). We found that B/B:B/b+b/b and B/B+B/b:b/b were significantly different in symptomatic patients compared to controls (p = 0.033 and p = 0.041, respectively). The VDR BsmI b/b genotype distribution was found to be lower in deceased patients than in living patients (p = 0.023). There was no significant difference between the groups in terms of TLR2 23bp indel genotype and allele distribution (p > 0.05). Our study results suggest that the VDR BsmI b allele may have a role in COVID-19 patients with symptomatic findings. These data need to be repeated in different ethnic and larger sample groups.
Collapse
Affiliation(s)
- Nilufer Kuruca
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Aynur Atilla
- Department of Infectious Disease and Clinical Microbiology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Muhammed Taha Kaya
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Sedat Gokmen
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Kastamonu University, Kastamonu, Turkey
| | - Ayse Feyda Nursal
- Department of Medical Genetics, Faculty of Medicine, Hitit University, Corum, Turkey
| | - Ozgur Kilic
- Department of Internal Medicine, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Tuba Kuruoglu
- Department of Infectious Disease and Clinical Microbiology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Fatih Temocin
- Department of Infectious Disease and Clinical Microbiology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Tolga Guvenc
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Serbulent Yigit
- Department of Genetics, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Dilek Guvenc
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| |
Collapse
|
2
|
Alhabibi AM, Hassan AS, Abd Elbaky NM, Eid HA, Khalifa MAAA, Wahab MA, Althoqapy AA, Abdou AE, Zakaria DM, Nassef EM, Kasim SA, Saleh OI, Elsheikh AA, Lotfy M, Sayed A. Impact of Toll-Like Receptor 2 and 9 Gene Polymorphisms on COVID-19: Susceptibility, Severity, and Thrombosis. J Inflamm Res 2023; 16:665-675. [PMID: 36825132 PMCID: PMC9942505 DOI: 10.2147/jir.s394927] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/26/2023] [Indexed: 02/18/2023] Open
Abstract
Background Toll-like receptors (TLRs) play an important role in activation of innate and adaptive immune responses. Aim We aimed to detect the association between TLR2 rs5743708 G>A and TLR9 rs5743836 C>T variants and COVID-19 disease susceptibility, severity, and thrombosis by using neutrophil extracellular traps (NETs). Subjects and Methods We included 100 adult COVID-19 patients as well as 100 age- and gender-matched normal controls. Participants were genotyped for TLR2 rs5743708 and TLR9 rs5743836. Citrullinated Histone (H3) was detected as an indicator of NETs. Results The mutant (G/A and C/C) genotypes and (A and C) alleles of TLR2 rs5743708 and TLR9 rs5743836, respectively, have been significantly related to a higher risk of COVID-19 infection, representing a significant risk factor for the severity of COVID-19. There was no significant association between the two variants and citrullinated histone (H3). Conclusion TLR2 rs5743708 and TLR9 rs5743836 variants have been significantly related to a higher risk and severity of COVID-19 infection but had no effect on thrombus formation.
Collapse
Affiliation(s)
- Alshaymaa M Alhabibi
- Departments of Clinical Pathology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt,Correspondence: Alshaymaa M Alhabibi, Tel +201002894075, Email
| | - Asmaa S Hassan
- Departments of Clinical Pathology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | | | - Hoda Asaad Eid
- Chest Disease, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | | | - Maisa A Wahab
- Vascular Surgery, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Azza Ali Althoqapy
- Medical Microbiology and Immunology Department, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Aml E Abdou
- Medical Microbiology and Immunology Department, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | | | - Eman Mostafa Nassef
- Internal Medicine, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Sammar Ahmed Kasim
- Internal Medicine, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Ola I Saleh
- Radio-Diagnosis, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Asmaa Abdelghany Elsheikh
- Community and Occupational Medicine, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Mahmoud Lotfy
- Molecular Biology Department, Genetic Engineering & Biotechnology Research Institute, University of Sadat City, Sadat City, Minufiya, Egypt
| | - Alaa Sayed
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| |
Collapse
|
3
|
Guillamat-Prats R. Role of Mesenchymal Stem/Stromal Cells in Coagulation. Int J Mol Sci 2022; 23:ijms231810393. [PMID: 36142297 PMCID: PMC9499599 DOI: 10.3390/ijms231810393] [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: 08/09/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/23/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are widely used in disease models in order to control several phases in the response to injuries, immune reaction, wound healing, and regeneration. MSCs can act upon both the innate and adaptive immune systems and target a broad number of functions, such as the secretion of cytokines, proteolytic enzymes, angiogenic factors, and the regulating of cell proliferation and survival. The role of MSCs in coagulation has been less studied. This review evaluates the properties and main functions of MSCs in coagulation. MSCs can regulate coagulation in a wide range of pathways. MSCs express and release tissue factors (TF), one of the key regulators of the extrinsic coagulation pathways; MSCs can trigger platelet production and contribute to platelet activation. Altogether, MSCs seem to have a pro-thrombotic role and their superior characterization prior to their administration is necessary in order to prevent adverse coagulation events.
Collapse
Affiliation(s)
- Raquel Guillamat-Prats
- Lung Immunity Translational Research Group in Respiratory Diseases, Germans Trias i Pujol Research Institute (IGTP), 08914 Badalona, Spain
| |
Collapse
|
4
|
Jahn K, Kohler TP, Swiatek LS, Wiebe S, Hammerschmidt S. Platelets, Bacterial Adhesins and the Pneumococcus. Cells 2022; 11:cells11071121. [PMID: 35406684 PMCID: PMC8997422 DOI: 10.3390/cells11071121] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 01/25/2023] Open
Abstract
Systemic infections with pathogenic or facultative pathogenic bacteria are associated with activation and aggregation of platelets leading to thrombocytopenia and activation of the clotting system. Bacterial proteins leading to platelet activation and aggregation have been identified, and while platelet receptors are recognized, induced signal transduction cascades are still often unknown. In addition to proteinaceous adhesins, pathogenic bacteria such as Staphylococcus aureus and Streptococcus pneumoniae also produce toxins such as pneumolysin and alpha-hemolysin. They bind to cellular receptors or form pores, which can result in disturbance of physiological functions of platelets. Here, we discuss the bacteria-platelet interplay in the context of adhesin–receptor interactions and platelet-activating bacterial proteins, with a main emphasis on S. aureus and S. pneumoniae. More importantly, we summarize recent findings of how S. aureus toxins and the pore-forming toxin pneumolysin of S. pneumoniae interfere with platelet function. Finally, the relevance of platelet dysfunction due to killing by toxins and potential treatment interventions protecting platelets against cell death are summarized.
Collapse
|
5
|
Alzahrani B, Gaballa MMS, Tantawy AA, Moussa MA, Shoulah SA, Elshafae SM. Blocking Toll-like receptor 9 attenuates bleomycin-induced pulmonary injury. J Pathol Transl Med 2022; 56:81-91. [PMID: 35220710 PMCID: PMC8934996 DOI: 10.4132/jptm.2021.12.27] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/27/2021] [Indexed: 11/17/2022] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) is one of the most common complications in coronavirus disease 2019 patients suffering from acute lung injury (ALI). In ARDS, marked distortion of pulmonary architecture has been reported. The pulmonary lesions in ARDS include hemodynamic derangements (such as alveolar edema and hemorrhage), vascular and bronchiolar damage, interstitial inflammatory cellular aggregations, and eventually fibrosis. Bleomycin induces ARDS-representative pulmonary damage in mice and rats; therefore, we used bleomycin model mice in our study. Recently, Toll-like receptor 9 (TLR9) was implicated in the development of ARDS and ALI. Methods In this study, we evaluated the efficiency of a TLR9 blocker (ODN2088) on bleomycin-induced pulmonary damage. We measured the apoptosis rate, inflammatory reaction, and fibroplasia in bleomycin- and bleomycin + ODN2088-treated mice. Results Our results showed a significant amelioration in bleomycin-induced damage to pulmonary architecture following ODN2088 treatment. A marked decrease in pulmonary epithelial and endothelial apoptosis rate as measured by cleaved caspase-3 expression, inflammatory reaction as indicated by tumor necrosis factor α expression, and pulmonary fibrosis as demonstrated by Van Gieson staining and α-smooth muscle actin immunohistochemistry were observed following ODN2088 treatment. Conclusions All these findings indicate that blocking downstream TLR9 signaling could be beneficial in prevention or mitigation of ARDS through hemodynamic derangements, inflammation, apoptosis, and fibrosis.
Collapse
Affiliation(s)
- Badr Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Mohamed M S Gaballa
- Department of Pathology, Faculty of Veterinary Medicine, Benha University, Tukh, Egypt
| | - Ahmed A Tantawy
- Department of Pathology, Faculty of Veterinary Medicine, Benha University, Tukh, Egypt
| | - Maha A Moussa
- Department of Statistics, Faculty of Commerce, Benha University, Benha, Egypt
| | - Salma A Shoulah
- Department of Animal Medicine (Infectious Diseases), Faculty of Veterinary Medicine, Benha University, Tukh, Egypt
| | - Said M Elshafae
- Department of Pathology, Faculty of Veterinary Medicine, Benha University, Tukh, Egypt
| |
Collapse
|
6
|
Single Nucleotide Polymorphisms from CSF2, FLT1, TFPI and TLR9 Genes Are Associated with Prelabor Rupture of Membranes. Genes (Basel) 2021; 12:genes12111725. [PMID: 34828331 PMCID: PMC8620696 DOI: 10.3390/genes12111725] [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: 09/25/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022] Open
Abstract
A prelabor rupture of membranes (PROM) and its subtypes, preterm PROM (pPROM) and term PROM (tPROM), are associated with disturbances in the hemostatic system and angiogenesis. This study was designed to demonstrate the role of single nucleotide polymorphisms (SNPs), localized in CSF2 (rs25881), FLT1 (rs722503), TFPI (C-399T) and TLR9 (rs352140) genes, in PROM. A population of 360 women with singleton pregnancy consisted of 180 PROM cases and 180 healthy controls. A single-SNP analysis showed a similar distribution of genotypes in the studied polymorphisms between the PROM or the pPROM women and the healthy controls. Double-SNP TT variants for CSF2 and FLT1 polymorphisms, CC variants for TLR9 and TFPI SNPs, TTC for CSF2, FLT1 and TLR9 polymorphisms, TTT for FLT1, TLR9 and TFPI SNPs and CCCC and TTTC complex variants for all tested SNPs correlated with an increased risk of PROM after adjusting for APTT, PLT parameters and/or pregnancy disorders. The TCT variants for the CSF2, FLT1 and TLR9 SNPs and the CCTC for the CSF2, FLT1, TLR9 and TFPI polymorphisms correlated with a reduced risk of PROM when corrected by PLT and APTT, respectively. We concluded that the polymorphisms of genes, involved in hemostasis and angiogenesis, contributed to PROM.
Collapse
|
7
|
Sha S, Pearson JA, Peng J, Hu Y, Huang J, Xing Y, Zhang L, Zhu Y, Zhao H, Wong FS, Chen L, Wen L. TLR9 Deficiency in B Cells Promotes Immune Tolerance via Interleukin-10 in a Type 1 Diabetes Mouse Model. Diabetes 2021; 70:504-515. [PMID: 33154070 PMCID: PMC7881860 DOI: 10.2337/db20-0373] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 11/01/2020] [Indexed: 12/22/2022]
Abstract
Toll-like receptor 9 (TLR9) is highly expressed in B cells, and B cells are important in the pathogenesis of type 1 diabetes (T1D) development. However, the intrinsic effect of TLR9 in B cells on β-cell autoimmunity is not known. To fill this knowledge gap, we generated NOD mice with a B-cell-specific deficiency of TLR9 (TLR9fl/fl/CD19-Cre+ NOD). The B-cell-specific deletion of TLR9 resulted in near-complete protection from T1D development. Diabetes protection was accompanied by an increased proportion of interleukin-10 (IL-10)-producing B cells. We also found that TLR9-deficient B cells were hyporesponsive to both innate and adaptive immune stimuli. This suggested that TLR9 in B cells modulates T1D susceptibility in NOD mice by changing the frequency and function of IL-10-producing B cells. Molecular analysis revealed a network of TLR9 with matrix metalloproteinases, tissue inhibitor of metalloproteinase-1, and CD40, all of which are interconnected with IL-10. Our study has highlighted an important connection of an innate immune molecule in B cells to the immunopathogenesis of T1D. Thus, targeting the TLR9 pathway, specifically in B cells, may provide a novel therapeutic strategy for T1D treatment.
Collapse
Affiliation(s)
- Sha Sha
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
- Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, Shandong, China
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
| | - James A Pearson
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
| | - Jian Peng
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
| | - Youjia Hu
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
| | - Juan Huang
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
| | - Yanpeng Xing
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
- Department of Gastrointestinal Surgery, First Hospital of Jilin University, Changchun, Jilin, China
| | - Luyao Zhang
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
- Department of Gastrointestinal Surgery, First Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Zhu
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT
| | - Hongyu Zhao
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT
| | - F Susan Wong
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
| | - Li Chen
- Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, Shandong, China
| | - Li Wen
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
| |
Collapse
|
8
|
Bezemer GFG, Garssen J. TLR9 and COVID-19: A Multidisciplinary Theory of a Multifaceted Therapeutic Target. Front Pharmacol 2021; 11:601685. [PMID: 33519463 PMCID: PMC7844586 DOI: 10.3389/fphar.2020.601685] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
By mapping the clinical pathophysiology of the novel coronavirus disease 2019 (COVID-19) against insights from virology, immunology, genomics, epidemiology and pharmacology, it is here proposed that the pathogen recognition receptor called toll like receptor 9 (TLR9) might have a pivotal role in the pathogenesis of COVID-19. Severe Acute Respiratory Syndrome Coronavirus 2, is causing the greatest global social and economic disruption since world war II. Lack of a vaccine, lack of successful treatment and limitations of the healthcare workforce and resources needed to safeguard patients with severe COVID-19 on the edge of life, demands radical preventive measures. It is urgently needed to identify biomarkers and drug candidates so that vulnerable individuals can be recognized early and severe multi-organ complications can be prevented or dampened. The TLR9 COVID-19 hypothesis describes a mechanism of action that could explain a wide spectrum of manifestations observed in patients with severe COVID-19. The introduced hypothesis proposes biomarkers for identification of vulnerable individuals and positions TLR9 as a promising multifaceted intervention target for prevention and/or treatment of COVID-19. TLR9 agonists might have value as prophylactic vaccine adjuvants and therapeutic immune stimulators at the early onset of disease. Additionally, in this current manuscript it is proposed for the first time that TLR9 could be considered as a target of "inhibition" aimed to dampen hyperinflammation and thrombotic complications in vulnerable patients that are at risk of developing late stages of COVID-19. The readily availability of TLR9 modulating drug candidates that have reached clinical testing for other disorders could favor a fast track development scenario, an important advantage under the current high unmet medical need circumstances regarding COVID-19.
Collapse
Affiliation(s)
- Gillina F. G. Bezemer
- Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Impact Station, Hilversum, Netherlands
| | - Johan Garssen
- Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Department of Immunology, Nutricia Research BV, Utrecht, Netherlands
| |
Collapse
|
9
|
Cheng Y, Liu B, Qian H, Yang H, Wang Y, Wu Y, Shen F. BAY11-7082 inhibits the expression of tissue factor and plasminogen activator inhibitor-1 in type-II alveolar epithelial cells following TNF-α stimulation via the NF-κB pathway. Exp Ther Med 2020; 21:177. [PMID: 33552241 DOI: 10.3892/etm.2020.9608] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/17/2020] [Indexed: 12/26/2022] Open
Abstract
Pulmonary inflammation strongly promotes alveolar hypercoagulation and fibrinolytic inhibition. NF-κB signaling regulates the expression of molecules associated with coagulation and fibrinolytic inhibition in type-II alveolar epithelial cells (AECII) stimulated by lipopolysaccharide. However, whether TNF-α-induced alveolar hypercoagulation and fibrinolysis inhibition is also associated with the NF-κB pathway remains to be determined. The aim of the present study was to determine whether BAY11-7082, an inhibitor of the NF-κB pathway, inhibits the expressions of tissue factor (TF) and plasminogen activator inhibitor-1 (PAI-1) in AECⅡ in response to TNF-α. Rat AECII were treated with BAY11-7082 for 24 h and stimulated with TNF-α for 1 h. The expression of TF and PAI-1 were determined using western blotting and reverse transcription-quantitative PCR. The concentrations of TF and PAI-1 in culture supernatant were also measured by ELISA. Moreover, levels of NF-κB p65 (p65), phosphorylated (p)-p65 (p-p65), inhibitor of NF-κB α (IκBα) and p-IκBα were also evaluated. Immunofluorescence was used to detect p65 levels in cell nuclei. TNF-α significantly promoted TF and PAI-1 expression either at the mRNA or protein level in AECII cells. Concentrations of TF and PAI-1 in supernatant also significantly increased upon TNF-α stimulation. Furthermore, TNF-α upregulated the levels of p-IκBα, p65, and p-p65 in the cytoplasm. Immunofluorescence analysis indicated that TNF-α increased p65 translocation from the cytoplasm to the nucleus. However, AECII pre-treated with BAY11-7082 expressed lower levels of TF and PAI-1 following TNF-α treatment. Levels of p-IκBα, p65 and p-p65 in the cytoplasm also decreased, and translocation of p65 from cytoplasm into the nucleus was inhibited by BAY11-7082 pretreatment. These findings suggest that BAY11-7082 improves the hypercoagulation and fibrinolytic inhibition induced by TNF-α in alveolar epithelial cells via the NF-κB signaling pathway. BAY11-7082 might represent a therapeutic option for alveolar hypercoagulation and fibrinolytic inhibition in acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Yumei Cheng
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Bo Liu
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Hong Qian
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Huilin Yang
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Yahui Wang
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Yanqi Wu
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Feng Shen
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| |
Collapse
|
10
|
Pan YK, Li CF, Gao Y, Wang YC, Sun XQ. Effect of miR-27b-5p on apoptosis of human vascular endothelial cells induced by simulated microgravity. Apoptosis 2020; 25:73-91. [PMID: 31768842 PMCID: PMC6965346 DOI: 10.1007/s10495-019-01580-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Weightlessness-induced cardiovascular dysfunction can lead to physiological and pathological consequences. It has been shown that spaceflight or simulated microgravity can alter expression profiles of some microRNAs (miRNAs). Here, we attempt to identify the role of miRNAs in human umbilical vein endothelial cells (HUVECs) apoptosis under simulated microgravity. RNA-sequencing and quantitative real-time PCR (qRT-PCR) assays were used to identify differentially expressed miRNAs in HUVECs under simulated microgravity. Then we obtained the target genes of these miRNAs through target analysis software. Moreover, GO and KEGG enrichment analysis were performed. The effects of these miRNAs on HUVECs apoptosis were evaluated by flow cytometry, Western blot and Hoechst staining. Furthermore, we obtained the target gene of miR-27b-5p by luciferase assay, qRT-PCR and Western blot. Finally, we investigated the relationship between this target gene and miR-27b-5p in HUVECs apoptosis under normal gravity or simulated microgravity. We found 29 differentially expressed miRNAs in HUVECs under simulated microgravity. Of them, the expressions of 3 miRNAs were validated by qRT-PCR. We demonstrated that miR-27b-5p affected HUVECs apoptosis by inhibiting zinc fingers and homeoboxes 1 (ZHX1). Our results reported here demonstrate for the first time that simulated microgravity can alter the expression of some miRNAs in HUVECs and miR-27b-5p may protect HUVECs from apoptosis under simulated microgravity by targeting ZHX1.
Collapse
Affiliation(s)
- Yi-Kai Pan
- School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
| | - Cheng-Fei Li
- Key Lab of Aerospace Medicine, Chinese Ministry of Education, Xi'an, 710032, Shaanxi, China
| | - Yuan Gao
- School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
| | - Yong-Chun Wang
- Key Lab of Aerospace Medicine, Chinese Ministry of Education, Xi'an, 710032, Shaanxi, China.
| | - Xi-Qing Sun
- School of Aerospace Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
11
|
Bhagwani A, Thompson AAR, Farkas L. When Innate Immunity Meets Angiogenesis-The Role of Toll-Like Receptors in Endothelial Cells and Pulmonary Hypertension. Front Med (Lausanne) 2020; 7:352. [PMID: 32850883 PMCID: PMC7410919 DOI: 10.3389/fmed.2020.00352] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/12/2020] [Indexed: 01/16/2023] Open
Abstract
Toll-like receptors serve a central role in innate immunity, but they can also modulate cell function in various non-immune cell types including endothelial cells. Endothelial cells are necessary for the organized function of the vascular system, and part of their fundamental role is also the regulation of immune function and inflammation. In this review, we summarize the current knowledge of how Toll-like receptors contribute to the immune and non-immune functions of the endothelial cells.
Collapse
Affiliation(s)
- Aneel Bhagwani
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH, United States
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, United States
| | - A. A. Roger Thompson
- Department of Infection, Immunity & Cardiovascular Disease, Faculty of Medicine, Dentistry & Health, University of Sheffield, Sheffield, United Kingdom
| | - Laszlo Farkas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
12
|
Saito M, Makino Y, Inoue K, Watanabe Y, Hoshi O, Kubota T. Anti-DNA antibodies cross-reactive with β 2-glycoprotein I induce monocyte tissue factor through the TLR9 pathway. Immunol Med 2020; 44:124-135. [PMID: 32701417 DOI: 10.1080/25785826.2020.1796285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Antibodies specific for cardiolipin (CL)-β2-glycoprotein I (β2GPI) are known to induce tissue factor (TF) expression by monocytes and endothelial cells which leads to a prothrombotic state in antiphospholipid syndrome (APS), but the mechanism is not fully elucidated. Previously, we reported that the mouse monoclonal anti-CL-β2GPI antibody WB-6 cross-reacts with DNA, enters monocytes via binding to cell surface DNA, and induces TF expression. The current study aimed to identify the intracellular signaling pathways involved in this process. The binding of WB-6 to CL-β2GPI or DNA, and endocytosis was not prevented by chloroquine, but pre-treatment of the cells with chloroquine significantly suppressed TF expression. TLR9 inhibitory oligodeoxynucleotide also suppressed the WB-6-induced TF expression, suggesting a pivotal role of the TLR9 pathway in TF production. Serum antibodies obtained from a patient with APS accompanying systemic lupus erythematosus (SLE) bound to both CL-β2GPI and DNA, and induced TF in normal monocytes. This effect was suppressed by chloroquine, and abolished by removal of the DNA-binding activity. These results suggest that induction of TF expression results from TLR9 activation by DNA which was internalized together with cross-reactive antibodies produced in secondary APS accompanying SLE.
Collapse
Affiliation(s)
- Masumi Saito
- Department of Immunopathology, Tokyo Medical and Dental University (TMDU) Graduate School of Medical and Dental Sciences, Tokyo, Japan.,Laboratory for Clinical Research, Nippon Medical School, Tokyo, Japan
| | - Yumi Makino
- Department of Immunopathology, Tokyo Medical and Dental University (TMDU) Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Kumi Inoue
- Department of Immunopathology, Tokyo Medical and Dental University (TMDU) Graduate School of Medical and Dental Sciences, Tokyo, Japan.,Department of Anatomical and Physiological Science, TMDU Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Yoshino Watanabe
- Department of Immunopathology, Tokyo Medical and Dental University (TMDU) Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Osamu Hoshi
- Department of Anatomical and Physiological Science, TMDU Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Tetsuo Kubota
- Department of Immunopathology, Tokyo Medical and Dental University (TMDU) Graduate School of Medical and Dental Sciences, Tokyo, Japan.,Department of Anatomical and Physiological Science, TMDU Graduate School of Medical and Dental Sciences, Tokyo, Japan.,Department of Medical Technology, Tsukuba International University, Ibaraki, Japan
| |
Collapse
|
13
|
Shepard CR. TLR9 in MAFLD and NASH: At the Intersection of Inflammation and Metabolism. Front Endocrinol (Lausanne) 2020; 11:613639. [PMID: 33584545 PMCID: PMC7880160 DOI: 10.3389/fendo.2020.613639] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/10/2020] [Indexed: 12/15/2022] Open
Abstract
Toll-Like Receptor 9 (TLR9) is an ancient receptor integral to the primordial functions of inflammation and metabolism. TLR9 functions to regulate homeostasis in a healthy system under acute stress. The literature supports that overactivation of TLR9 under the chronic stress of obesity is a critical driver of the pathogenesis of NASH and NASH-associated fibrosis. Research has focused on the core contributions of the parenchymal and non-parenchymal cells in the liver, adipose, and gut compartments. TLR9 is activated by endogenous circulating mitochondrial DNA (mtDNA). Chronically elevated circulating levels of mtDNA, caused by the stress of overnutrition, are observed in obesity, metabolic dysfunction-associated fatty liver disease (MAFLD), and NASH. Clinical evidence is supportive of TLR9 overactivation as a driver of disease. The role of TLR9 in metabolism and energy regulation may have an underappreciated contribution in the pathogenesis of NASH. Antagonism of TLR9 in NASH and NASH-associated fibrosis could be an effective therapeutic strategy to target both the inflammatory and metabolic components of such a complex disease.
Collapse
|
14
|
Wang G, Liu ZJ, Liu X, Liu FG, Li Y, Weng YB, Zhou JX. A study on the protective effects of CpG oligodeoxynucleotide-induced mucosal immunity against lung injury in a mouse acute respiratory distress syndrome model. J Cell Physiol 2019; 234:20118-20127. [PMID: 30953359 DOI: 10.1002/jcp.28613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/12/2019] [Accepted: 03/19/2019] [Indexed: 12/18/2022]
Abstract
This study aims to determine the feasibility of using oligodeoxynucleotides with unmethylated cytosine-guanine dinucleotide sequences (CpG ODN) as an immunity protection strategy for a mouse model of acute respiratory distress syndrome (ARDS). This is a prospective laboratory animal investigation. Twenty-week-old BALB/c mice in Animal research laboratory were randomized into groups. An ARDS model was induced in mice using lipopolysaccharides (LPSs). CpG ODN was intranasally and transrectally immunized before or after the 3rd and 7th days of establishing the ARDS model. Mice were euthanized on Day 7 after the second immunization. Then, retroorbital bleeding was carried out and the chest was rapidly opened to collect the trachea and tissues from both lungs for testing. CpG ODN significantly improved the pathologic impairment in mice lung, especially after the intranasal administration of 50 μg. This resulted in the least severe lung tissue injury. Furthermore, interleukin-6 (IL-6) and IL-8 concentrations were lower, which was second to mice treated with the rectal administration of 20 µg CpG ODN. In contrast, the nasal and rectal administration of CpG ODN in BALB/c mice before LPS immunization did not appear to exhibit any significant protective effects. The intranasal administration of CpG ODN may be a potential treatment approach to ARDS. More studies are needed to further determine the protective mechanism of CpG ODN.
Collapse
Affiliation(s)
- Guan Wang
- Department of Critical Care Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Critical Care Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Zong-Jian Liu
- Center Laboratory, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xuan Liu
- Center Laboratory, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Feng-Ge Liu
- Department of Critical Care Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yan Li
- Department of Critical Care Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yi-Bing Weng
- Department of Critical Care Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Jian-Xin Zhou
- Department of Critical Care Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Williams B, Neder J, Cui P, Suen A, Tanaka K, Zou L, Chao W. Toll-like receptors 2 and 7 mediate coagulation activation and coagulopathy in murine sepsis. J Thromb Haemost 2019; 17:1683-1693. [PMID: 31211901 PMCID: PMC7197442 DOI: 10.1111/jth.14543] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/10/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Sepsis is a life-threatening condition often manifested as marked inflammation and severe coagulopathy. Toll-like receptors (TLRs) play a pivotal role in inflammation, organ dysfunction and mortality in animal sepsis. OBJECTIVES To investigate the role of TLR signaling in mediating sepsis-induced coagulopathy (SIC) in a mouse model. METHODS Polymicrobial sepsis was created by cecal ligation and puncture (CLP) or fecal slurry peritoneal injection. To quantify global clotting function, two viscoelastic assays were performed with rotational thromboelastometry, and the results were presented as maximum clot firmness (MCF): (a) EXTEM to test tissue factor (TF)-initiated clot formation; and (b) FIBTEM to test EXTEM in the presence of a platelet inhibitor, cytochalasin D. Plasma coagulation factors were quantified with ELISA. TF gene expression and protein expression were determined with real-time quantitative reverse transcription PCR and flow cytometry, respectively. RESULTS Between 4 and 24 hours after CLP surgery, wild-type mice showed significant MCF reduction in both EXTEM and FIBTEM tests. This was accompanied by marked thrombocytopenia and a significant increase in the levels of plasminogen activator inhibitor-1, plasma TF, and D-dimer. In comparison, TLR2-/- and TLR7-/- CLP mice showed preserved MCF and platelet counts, and near-normal plasma TF levels. Bone marrow-derived macrophages treated with a TLR2 agonist Pam3cys-Ser-(Lys)4 (Pam3cys) or a TLR7 agonist (R837) showed marked increases in TF gene expression and protein expression. MicroRNA-146a, a newly identified proinflammatory mediator that is upregulated during sepsis, induced TF production via a TLR7-dependent mechanism. CONCLUSIONS Murine sepsis leads to an increased procoagulant response, thrombocytopenia, and global coagulopathy. TLR2 and TLR7 play an important role in procoagulant production and in SIC.
Collapse
Affiliation(s)
- Brittney Williams
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jessica Neder
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ping Cui
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Andrew Suen
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kenichi Tanaka
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Lin Zou
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
16
|
Liu B, Wang Y, Wu Y, Cheng Y, Qian H, Yang H, Shen F. IKKβ regulates the expression of coagulation and fibrinolysis factors through the NF-κB canonical pathway in LPS-stimulated alveolar epithelial cells type II. Exp Ther Med 2019; 18:2859-2866. [PMID: 31572531 PMCID: PMC6755483 DOI: 10.3892/etm.2019.7928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 07/12/2019] [Indexed: 01/11/2023] Open
Abstract
Aim: Hypercoagulation and fibrinolysis inhibition in the alveolar cavity are important characteristics in acute respiratory distress syndrome (ARDS). Alveolar epithelial cells type II (AEC II) have been confirmed to have significant role in regulating alveolar hypercoagulation and fibrinolysis inhibition, but the mechanism is unknown. Nuclear factor-κB (NF-κB) signaling pathway has been demonstrated to participate in the pathogenesis of these two abnormalities in ARDS. The purpose of the present study is to explore whether controlling the upstream crucial factor IκB kinase (IKK)β could regulate coagulation and fibrinolysis factors in LPS-stimulated AEC II. Materials and methods: An IKKβ gene regulation model (IKKβ+/+ and IKKβ−/−) was prepared using lentiviral vector transfection. The models with wild type cells were all stimulated by lipopolysaccharide (LPS) or saline for 24 h. Expression of the related proteins were determined by western-blotting, ELISA and revere transcription-PCR respectively. Tissue factor (TF) procoagulant activity and nuclear p65 protein level were also detected. Results: IKKβ increased in IKKβ+/+ cells but decreased in IKKβ−/− cells. LPS stimulation promoted the expression of p-IκBα, p65, p-p65 and p-IKKβ as well as TF and plasminogen activator inhibitor (PAI)-1, at the mRNA or protein level, and this was significantly enhanced by IKKβ upregulation but weakened by IKKβ downregulation. TF procoagulant activity presented the same changes as the molecules above. ELISAs showed additional increases in the concentrations of as thrombin antithrombin, procollagen III propeptide, thrombomodulin and PAI-1 in IKKβ+/+ cell supernatant under LPS stimulation, however they decreased in IKKβ−/−. The level of as antithrombin III however, appeared to show the opposite change to those other factors. Immunofluorescence demonstrated a greatly enhanced expression of p65 in the nucleus by IKKβ upregulation, which was reduced by IKKβ downregulation. Conclusions: IKKβ could regulate the expression and secretion of coagulation and fibrinolysis factors in LPS-stimulated AEC II via the NF-κB p65 signaling pathway. The IKKβ molecule is expected to be a new target for prevention of coagulation and fibrinolysis abnormalities in ARDS.
Collapse
Affiliation(s)
- Bo Liu
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Yahui Wang
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Yanqi Wu
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Yumei Cheng
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Hong Qian
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Huilin Yang
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| | - Feng Shen
- Department of Critical Care Medicine, Guizhou Medical University Affiliated Hospital, Guiyang, Guizhou 550001, P.R. China
| |
Collapse
|
17
|
Liu B, Wu Y, Wang Y, Cheng Y, Yao L, Liu Y, Qian H, Yang H, Shen F. NF-κB p65 Knock-down inhibits TF, PAI-1 and promotes activated protein C production in lipopolysaccharide-stimulated alveolar epithelial cells type II. Exp Lung Res 2018; 44:241-251. [PMID: 30449218 DOI: 10.1080/01902148.2018.1505975] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Purpose/aim: Activated coagulation and reduced fibrinolysis in alveolar compartment are an important characteristics in acute respiratory distress syndrome (ARDS). Alveolar epithelial cell type II (AECII) participates in regulating the intra-alveolar abnormalities of coagulation and fibrinolysis mainly through adjusting the productions of tissue factor (TF), plasminogen activator inhibitor (PAI)-1 and activated protein C (APC) in ARDS. NF-κB signal pathway may be involved in coagulation regulation in sepsis-induced ALI. The purpose of this study was to testify the hypothesis that NF-κB p65 (p65) knock-down would improve the abnormalities of coagulation and fibrinolysis mediated by lipopolysaccharide (LPS) stimulation in AECII. MATERIALS AND METHODS p65 gene knock-down in AECII was achieved by small interfering RNA (siRNA) transfection. Rat AECII (RLE-6TN) with or without p65 gene knock-down were stimulated by LPS for 24 hours. And then cytolysate was used for TF, PAI-1 expression examination, and supernatant was collected for TF, PAI-1 and PC concentrations determination. Activation of NF-κB canonical pathway was simultaneously checked by western-blotting, RT-PCR and immunofluorescence respectively. RESULTS TF, PAI-1 expressions in normal cells obviously increased under LPS stimulation with NF-κB canonical pathway activation represented by high levels of p65, p-p65, p-IκB with increased nuclear translocation of p-p65. Cells with NF-κB p65 knock-down, however, showed significant decreases in TF, PAI-1, p65, p-p65, p-IκB expressions following LPS stimulation with significant reduction in p-p65 nuclear translocation as compared to normal and siRNA control cells. The high concentrations of TF, PAI-1 and low level of APC in supernatant induced by LPS in normal cells were significantly reversed through p65 knock-down. CONCLUSIONS The experimental findings demonstrate that NF-kB signaling pathway is involved in regulating the expressions of coagulation and fibrinolysis factors in LPS-stimulated AECII, which suggest that NF-kB signaling pathway may be a new target to correct intra-alveolar coagulation and fibrinolytic abnormalities in ARDS.
Collapse
Affiliation(s)
- Bo Liu
- a Department of Critical Care Medicine , The Affiliated Hospital of Guizhou Medical University , Guiyang , China
| | - Yanqi Wu
- a Department of Critical Care Medicine , The Affiliated Hospital of Guizhou Medical University , Guiyang , China
| | - Yahui Wang
- a Department of Critical Care Medicine , The Affiliated Hospital of Guizhou Medical University , Guiyang , China
| | - Yumei Cheng
- a Department of Critical Care Medicine , The Affiliated Hospital of Guizhou Medical University , Guiyang , China
| | - Ling Yao
- b Department of Critical Care Medicine , The Second Affiliated Hospital of Guizhou Medical University , Kaili China
| | - Yuqin Liu
- c Department of Critical Care Medicine , The Fourth People's Hospital of Zhenjiang Ctiy , Zhenjiang , China
| | - Hong Qian
- a Department of Critical Care Medicine , The Affiliated Hospital of Guizhou Medical University , Guiyang , China
| | - Huilin Yang
- a Department of Critical Care Medicine , The Affiliated Hospital of Guizhou Medical University , Guiyang , China
| | - Feng Shen
- a Department of Critical Care Medicine , The Affiliated Hospital of Guizhou Medical University , Guiyang , China
| |
Collapse
|
18
|
Yin Y, Liu X, Liu J, Cai E, Zhu H, Li H, Zhang L, Li P, Zhao Y. Beta-sitosterol and its derivatives repress lipopolysaccharide/d-galactosamine-induced acute hepatic injury by inhibiting the oxidation and inflammation in mice. Bioorg Med Chem Lett 2018; 28:1525-1533. [DOI: 10.1016/j.bmcl.2018.03.073] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/19/2018] [Accepted: 03/27/2018] [Indexed: 02/07/2023]
|
19
|
Boff D, Fagundes CT, Russo RC, Amaral FA. Innate Immunity and Inflammation: The Molecular Mechanisms Governing the Cross-Talk Between Innate Immune and Endothelial Cells. IMMUNOPHARMACOLOGY AND INFLAMMATION 2018:33-56. [DOI: 10.1007/978-3-319-77658-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
20
|
Sessile Innate Immune Cells. DAMAGE-ASSOCIATED MOLECULAR PATTERNS IN HUMAN DISEASES 2018. [PMCID: PMC7123606 DOI: 10.1007/978-3-319-78655-1_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this chapter, sessile cells of the innate immune system are briefly introduced. Defined as cells equipped with diverse pattern recognition molecules capable of detecting MAMPs and DAMPs, they encompass cells such as epithelial cells, fibroblasts, vascular cells, chondrocytes, osteoblasts, and adipocytes. Located at the body surfaces, epithelial cells represent the first line of innate immune defense against invading microbial pathogens. They are significant contributors to innate mucosal immunity and generate various antimicrobial defense mechanisms. Also, epithelial cells critically contribute to tissue repair via the phenomenon of re-epithelialization. Fibroblasts operate as classical sentinel cells of the innate immune system dedicated to responding to MAMPs and DAMPs emitted upon any tissue injury. Typically, fibroblasts synthesize most of the extracellular matrix of connective tissues, thereby playing a crucial role in tissue repair processes. Vascular cells of the innate immune system represent an evolutionarily developed first-line defense against any inciting insult hitting the vessel walls from the luminal side including bacteria, viruses, microbial toxins, and chemical noxa such as nicotine. Upon such insults and following recognition of MAMPs and DAMPs, vascular cells react with an innate immune response to create an acute inflammatory milieu in the vessel wall aimed at curing the vascular injury concerned. Chondrocytes, osteoblasts, and osteoclasts represent other vital cells of the skeletal system acting as cells of the innate immune system in its wider sense. These cells mediate injury-promoted DAMP-induced inflammatory and regenerative processes specific for the skeletal systems. Finally, adipocytes are regarded as highly active cells of the innate immune system. As white, brown, and beige adipocytes, they operate as a dynamic metabolic organ that can secrete certain bioactive molecules which have endocrine, paracrine, and autocrine actions.
Collapse
|
21
|
Foley JH, Conway EM. Cross Talk Pathways Between Coagulation and Inflammation. Circ Res 2017; 118:1392-408. [PMID: 27126649 DOI: 10.1161/circresaha.116.306853] [Citation(s) in RCA: 409] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 03/21/2016] [Indexed: 02/06/2023]
Abstract
Anatomic pathology studies performed over 150 years ago revealed that excessive activation of coagulation occurs in the setting of inflammation. However, it has taken over a century since these seminal observations were made to delineate the molecular mechanisms by which these systems interact and the extent to which they participate in the pathogenesis of multiple diseases. There is, in fact, extensive cross talk between coagulation and inflammation, whereby activation of one system may amplify activation of the other, a situation that, if unopposed, may result in tissue damage or even multiorgan failure. Characterizing the common triggers and pathways are key for the strategic design of effective therapeutic interventions. In this review, we highlight some of the key molecular interactions, some of which are already showing promise as therapeutic targets for inflammatory and thrombotic disorders.
Collapse
Affiliation(s)
- Jonathan H Foley
- From the Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom (J.H.F.); Katharine Dormandy Haemophilia Centre and Thrombosis Unit, Royal Free NHS Trust, London, United Kingdom (J.H.F.); and Centre for Blood Research, Department of Medicine, University of British Columbia, Vancouver, Canada (E.M.C.)
| | - Edward M Conway
- From the Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom (J.H.F.); Katharine Dormandy Haemophilia Centre and Thrombosis Unit, Royal Free NHS Trust, London, United Kingdom (J.H.F.); and Centre for Blood Research, Department of Medicine, University of British Columbia, Vancouver, Canada (E.M.C.).
| |
Collapse
|
22
|
Salvador B, Arranz A, Francisco S, Córdoba L, Punzón C, Llamas MÁ, Fresno M. Modulation of endothelial function by Toll like receptors. Pharmacol Res 2016; 108:46-56. [PMID: 27073018 DOI: 10.1016/j.phrs.2016.03.038] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 03/30/2016] [Accepted: 03/30/2016] [Indexed: 12/23/2022]
Abstract
Endothelial cells (EC) are able to actively control vascular permeability, coagulation, blood pressure and angiogenesis. Most recently, a role for endothelial cells in the immune response has been described. Therefore, the endothelium has a dual role controlling homeostasis but also being the first line for host defence and tissue damage repair thanks to its ability to mount an inflammatory response. Endothelial cells have been shown to express pattern-recognition receptors (PRR) including Toll-like receptors (TLR) that are activated in response to stimuli within the bloodstream including pathogens and damage signals. TLRs are strategic mediators of the immune response in endothelial cells but they also regulate the angiogenic process critical for tissue repair. Nevertheless, endothelial activation and angiogenesis can contribute to some pathologies. Thus, inappropriate endothelial activation, also known as endothelial dysfunction, through TLRs contributes to tissue damage during autoimmune and inflammatory diseases such as atherosclerosis, hypertension, ischemia and diabetes associated cardiovascular diseases. Also TLR induced angiogenesis is required for the growth of some tumors, atherosclerosis and rheumatoid arthritis, among others. In this review we discuss the importance of various TLRs in modulating the activation of endothelial cells and their importance in immunity to infection and vascular disease as well as their potential as therapeutic targets.
Collapse
Affiliation(s)
| | - Alicia Arranz
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, Madrid, Spain.
| | - Sara Francisco
- Diomune SL, Parque Científico de Madrid, Madrid, Spain; Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, Madrid, Spain.
| | - Laura Córdoba
- Diomune SL, Parque Científico de Madrid, Madrid, Spain.
| | - Carmen Punzón
- Diomune SL, Parque Científico de Madrid, Madrid, Spain.
| | | | - Manuel Fresno
- Diomune SL, Parque Científico de Madrid, Madrid, Spain; Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
23
|
Khakpour S, Wilhelmsen K, Hellman J. Vascular endothelial cell Toll-like receptor pathways in sepsis. Innate Immun 2015; 21:827-46. [DOI: 10.1177/1753425915606525] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/11/2015] [Indexed: 12/20/2022] Open
Abstract
The endothelium forms a vast network that dynamically regulates vascular barrier function, coagulation pathways and vasomotor tone. Microvascular endothelial cells are uniquely situated to play key roles during infection and injury, owing to their widespread distribution throughout the body and their constant interaction with circulating blood. While not viewed as classical immune cells, endothelial cells express innate immune receptors, including the Toll-like receptors (TLRs), which activate intracellular inflammatory pathways mediated through NF-κB and the MAP kinases. TLR agonists, including LPS and bacterial lipopeptides, directly upregulate microvascular endothelial cell expression of inflammatory mediators. Intriguingly, TLR activation also modulates microvascular endothelial cell permeability and the expression of coagulation pathway intermediaries. Microvascular thrombi have been hypothesized to trap microorganisms thereby limiting the spread of infection. However, dysregulated activation of endothelial inflammatory pathways is also believed to lead to coagulopathy and increased vascular permeability, which together promote sepsis-induced organ failure. This article reviews vascular endothelial cell innate immune pathways mediated through the TLRs as they pertain to sepsis, highlighting links between TLRs and coagulation and permeability pathways, and their role in healthy and pathologic responses to infection and sepsis.
Collapse
Affiliation(s)
- Samira Khakpour
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
- Biomedical Sciences and Immunology Programs, University of California, San Francisco, CA, USA
| | - Kevin Wilhelmsen
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
| | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
- Biomedical Sciences and Immunology Programs, University of California, San Francisco, CA, USA
| |
Collapse
|