1
|
Altered Moesin and Actin Cytoskeleton Protein Rearrangements Affect Transendothelial Permeability in Human Endothelial Cells upon Dengue Virus Infection and TNF-α Treatment. Viruses 2021; 13:v13102042. [PMID: 34696472 PMCID: PMC8537470 DOI: 10.3390/v13102042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
It has been hypothesized that the host, viral factors, and secreted cytokines (especially TNF-α) play roles in the pathogenesis of secondary dengue infections. Mass spectrometry-based proteomic screening of cytoskeleton fractions isolated from human endothelial (EA.hy926) cells upon dengue virus (DENV) infection and TNF-α treatment identified 450 differentially altered proteins. Among them, decreased levels of moesin, actin stress fiber rearrangements, and dot-like formations of vinculin were observed with western blot analyses and/or immunofluorescence staining (IFA). In vitro vascular permeability assays using EA.hy926 cells, seeded on collagen-coated transwell inserts, showed low levels of transendothelial electrical resistance in treated cells. The synergistic effects of DENV infection and TNF-α treatment caused cellular permeability changes in EA.hy926 cells, which coincided with decreasing moesin levels and the production of abnormal organizations of actin stress fibers and vinculin. Functional studies demonstrated moesin overexpression restored transendothelial permeability in DENV/TNF-α-treated EA.hy926 cells. The present study improves the understanding of the disruption mechanisms of cytoskeleton proteins in enhancing vascular permeability during DENV infection and TNF-α treatment. The study also suggests that these disruption mechanisms are major factors contributing to vascular leakage in severe dengue patients.
Collapse
|
2
|
Contribution of Ezrin on the Cell Surface Plasma Membrane Localization of Programmed Cell Death Ligand-1 in Human Choriocarcinoma JEG-3 Cells. Pharmaceuticals (Basel) 2021; 14:ph14100963. [PMID: 34681187 PMCID: PMC8540387 DOI: 10.3390/ph14100963] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/01/2021] [Accepted: 09/22/2021] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint blockade (ICB) antibodies targeting programmed cell death ligand-1 (PD-L1) and programmed cell death-1 (PD-1) have improved survival in patients with conventional single agent chemotherapy-resistant gestational trophoblastic neoplasia (GTN). However, many patients are resistant to ICB therapy, the mechanisms of which are poorly understood. Unraveling the regulatory mechanism for PD-L1 expression may provide a new strategy to improve ICB therapy in patients with GTN. Here, we investigated whether the ezrin/radixin/moesin (ERM) family, i.e., a group of scaffold proteins that crosslink actin cytoskeletons with several plasma membrane proteins, plays a role in the regulation of PD-L1 expression using JEG-3 cells, a representative human choriocarcinoma cell line. Our results demonstrate mRNA and protein expressions of ezrin, radixin, and PD-L1, as well as their colocalization in the plasma membrane. Intriguingly, immunoprecipitation experiments revealed that PD-L1 interacted with both ezrin and radixin and the actin cytoskeleton. Moreover, gene silencing of ezrin but not radixin strongly diminished the cell surface expression of PD-L1 without altering the mRNA level. These results indicate that ezrin may contribute to the cell surface localization of PD-L1 as a scaffold protein in JEG-3 cells, highlighting a potential therapeutic target to improve the current ICB therapy in GTN.
Collapse
|
3
|
Simó-Servat O, Ramos H, Bogdanov P, García-Ramírez M, Huerta J, Hernández C, Simó R. ERM Complex, a Therapeutic Target for Vascular Leakage Induced by Diabetes. Curr Med Chem 2021; 29:2189-2199. [PMID: 34042029 DOI: 10.2174/0929867328666210526114417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/30/2021] [Accepted: 05/06/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ezrin, radixin, and moesin (the ERM complex) interact directly with membrane proteins regulating their attachment to actin filaments. ERM protein activation modifies cytoskeleton organization and alters the endothelial barrier function, thus favoring vascular leakage. However, little is known regarding the role of ERM proteins in diabetic retinopathy (DR). OBJECTIVE This study aimed to examine whether overexpression of the ERM complex exists in db/db mice and its main regulating factors. METHOD 9 male db/db mice and 9 male db/+ aged 14 weeks were analyzed. ERM proteins were assessed by western blot and by immunohistochemistry. Vascular leakage was determined by the Evans blue method. To assess ERM regulation, HRECs were cultured in a medium containing 5.5 mM D-glucose (mimicking physiological conditions) and 25 mM D-glucose (mimicking hyperglycemia that occurs in diabetic patients). Moreover, treatment with TNF-α, IL-1β, or VEGF was added to a high glucose condition. The expression of ERM proteins was quantified by RT-PCR. Cell permeability was evaluated by measuring movements of FITC-dextran. RESULTS A significant increase of ERM in diabetic mice in comparison with non-diabetic mice was observed. A high glucose condition alone did not have any effect on ERM expression. However, TNF-α and IL-1β induced a significant increase in ERM proteins. CONCLUSIONS The increase of ERM proteins induced by diabetes could be one of the mechanisms involved in vascular leakage and could be considered as a therapeutic target. Moreover, the upregulation of the ERM complex by diabetes is induced by inflammatory mediators rather than by high glucose itself.
Collapse
Affiliation(s)
- Olga Simó-Servat
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Hugo Ramos
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Patricia Bogdanov
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Marta García-Ramírez
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Jordi Huerta
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Cristina Hernández
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Rafael Simó
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| |
Collapse
|
4
|
Moesin Is a Novel Biomarker of Endothelial Injury in Sepsis. J Immunol Res 2021; 2021:6695679. [PMID: 33628853 PMCID: PMC7896848 DOI: 10.1155/2021/6695679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/19/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023] Open
Abstract
Objective Increased vascular permeability and inflammation are principal hallmark of sepsis. Moesin (MSN) is a membrane-associated cytoskeleton protein and crucial for the vascular endothelial function. This study is aimed at evaluating the role of MSN in endothelial injury during the process of sepsis. Methods Serum MSN in septic patients was measured by ELISA. BALB/c mice were injected with different doses of lipopolysaccharide (LPS) or underwent cecal ligation and single or double puncture (CLP) to mimic sublethal and lethal sepsis. After treatment, their serum MSN and PCT levels, wet to dry lung weights (W/D ratio), bronchoalveolar lavage fluid (BALF) protein concentrations, and lung injury scores were measured. The impact of MSN silencing on LPS-altered Rock1/myosin light chain (MLC), NF-κB, and inflammatory factors in human microvascular endothelial cells (HMECs), as well as monolayer HMEC permeability, was tested in vitro. Results Compared with healthy controls, serum MSN increased in septic patients and was positively correlated with SOFA scores and serum PCT levels in septic patients. LPS injection significantly increased serum the MSN and PCT expression, BALF protein levels, and W/D ratio, and the serum MSN levels were positively correlated with serum PCT, lung W/D ratio, and lung injury scores in mice. Similar results were obtained in the way of CLP modelling. LPS enhanced MSN, MLC, NF-κB phosphorylation, increased Rock1 expression, and inflammatory factors release in the cultured HMECs, while MSN silencing significantly mitigated the LPS-induced Rock1 and inflammatory factor expression, NF-κB, and MLC phosphorylation as well as the monolayer hyperpermeability in HMECs. Conclusions Increased serum MSN contributes to the sepsis-related endothelium damages by activating the Rock1/MLC and NF-κB signaling and may be a potential biomarker for evaluating the severity of sepsis.
Collapse
|
5
|
Simó-Servat O, Hernández C, Simó R. The ERM Complex: A New Player Involved in Diabetes-induced Vascular Leakage. Curr Med Chem 2020; 27:3012-3022. [PMID: 30332939 DOI: 10.2174/0929867325666181016162327] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/27/2018] [Accepted: 10/09/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Microvascular complications remain an important cause of morbidity in diabetic patients, and they are associated with a significant economic burden for healthcare systems. Vascular leakage is one of the earlier hallmarks in diabetic microvascular complications. Ezrin, Radixin and Moesin (ERM) proteins have recently been involved in vascular dysfunction under the effect of molecular mediators of diabetes complications. In this review, we will present the available evidence regarding the role of these proteins in vascular leakage and their putative implication in diabetic microvascular complications. METHODS AND RESULTS A comprehensive literature search of the electronic MEDLINE database was performed between November 2017 and January 2018. As a result, 36 articles have been reviewed and discussed. DISCUSSION ERM proteins are cytoskeleton-membrane linkers, and when activated in endothelial cells are able to induce cytoskeleton reorganization in stress fibers leading to the disassembly of focal adhesions and the formation of paracellular gaps which result in an increase of vascular permeability. The activation of these proteins is induced by mediators involved in diabetic complications such as PKC activation, TNF-α, AGEs and oxidative stress. In conclusion, ERMs play an essential role in endothelium homeostasis and can be envisaged as a new therapeutic molecular target for preventing or arresting diabetes-induced vascular leakage.
Collapse
Affiliation(s)
- Olga Simó-Servat
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Barcelona, Spain.,Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ICSIII), Madrid, Spain
| | - Cristina Hernández
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Barcelona, Spain.,Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ICSIII), Madrid, Spain
| | - Rafael Simó
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Barcelona, Spain.,Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ICSIII), Madrid, Spain
| |
Collapse
|
6
|
Jeong SY, Kim J, Park EK, Baek MC, Bae JS. Inhibitory functions of maslinic acid on particulate matter-induced lung injury through TLR4-mTOR-autophagy pathways. ENVIRONMENTAL RESEARCH 2020; 183:109230. [PMID: 32058145 DOI: 10.1016/j.envres.2020.109230] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/30/2020] [Accepted: 02/04/2020] [Indexed: 06/10/2023]
Abstract
Particulate matter (PM), the collection of all liquid and solid particles suspended in air, includes both organic and inorganic particles, many of which are health-hazards. PM particles with a diameter equal to or less than 2.5 μm (PM2.5) is a form of air pollutant that causes significant lung damage when inhaled. Maslinic acid (MA) prevents oxidative stress and pro-inflammatory cytokine generation, but there is little information available regarding its role in PM-induced lung injury. Therefore, the purpose of this study was to determine the protective activity of MA against PM2.5-induced lung injury. The mice were divided into seven groups (n = 10 each): a mock control group, an MA control (0.8 mg/kg mouse body weight) group, an opted PM2.5 produced from diesel (10 mg/kg mouse body weight) group, a diesel PM2.5+MA (0.2, 0.4, 0.6, and 0.8 mg/kg mouse body weight) groups. Mice were treated with MA via tail-vein injection 30 min after the intratracheal instillation of a diesel PM2.5. Changes in the wet/dry weight ratio of the lung tissue, total protein/total cell and lymphocyte counts, inflammatory cytokines in the bronchoalveolar lavage fluid (BALF), vascular permeability, and histology were monitored in diesel PM2.5-treated mice. The results showed that MA reduced pathological lung injury, the wet/dry weight ratio of the lung tissue, and hyperpermeability caused by diesel PM2.5. MA also inhibited diesel PM2.5-induced myeloperoxidase (MPO) activity in the lung tissue, decreased the levels of diesel PM2.5-induced inflammatory cytokines, including tumor necrosis factor (TNF)-α and interleukin (IL)-1β, reduced nitric oxide (NO) and total protein in the BALF, and effectively attenuated diesel PM2.5-induced increases in the number of lymphocytes in the BALF. In addition, MA increased the protein phosphorylation of the mammalian target of rapamycin (mTOR) and dramatically suppressed diesel PM2.5-stimulated expression of toll-like receptor 4 (TLR4), MyD88, and the autophagy-related proteins LC3 II and Beclin 1. In conclusion, these findings indicate that MA has a critical anti-inflammatory effect due to its ability to regulate both the TLR4-MyD88 and mTOR-autophagy pathways and may thus be a potential therapeutic agent against diesel PM2.5-induced lung injury.
Collapse
Affiliation(s)
- So Yeon Jeong
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Jaehong Kim
- Department of Biochemistry, College of Medicine, Gachon University, Incheon, 21999, Republic of Korea
| | - Eui Kyun Park
- Department of Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu, 41940, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
7
|
Kong Q, Li Y, Liang Q, Xie J, Li X, Fang J. SIRT6-PARP1 is involved in HMGB1 polyADP-ribosylation and acetylation and promotes chemotherapy-induced autophagy in leukemia. Cancer Biol Ther 2020; 21:320-331. [PMID: 31928132 DOI: 10.1080/15384047.2019.1702397] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
High mobility group box protein 1 (HMGB1) is an evolutionarily conserved non-histone chromatin-binding protein. In a previous study, we showed that treating leukemic cells with chemotherapeutic drugs leads to the translocation of HMGB1, which is involved in autophagy and ultimately promotes chemoresistance in leukemia. However, the underlying translocation mechanism of HMGB1 in chemotherapy-induced autophagy remains unclear. In this study, we showed that knockdown of SIRT6 or PARP1 gene expression significantly inhibited HMGB1 cytoplasmic translocation and autophagy. Meanwhile, we found that SIRT6, an important upstream protein of PARP1, associated with PARP1, leading to the stimulation of polyADP-ribose polymerase activity. We further demonstrated that SIRT6 and PARP1 activation were required for chemotherapy-induced ADP-ribosylation of HMGB1 in leukemic cells and then influenced the acetylation of HMGB1, finally promoting the autophagy of leukemic cells mediated by HMGB1 translocation. These findings provide new insights into the mechanism of chemotherapeutic drug resistance. Targeting the HMGB1 translocation may overcome autophagy-related chemoresistance in leukemia.
Collapse
Affiliation(s)
- Qian Kong
- Department of Pediatrics, TheThird Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P.R. China
| | - Yunyao Li
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P.R. China.,Department of Pediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangdong, P.R. China
| | - Qixiang Liang
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P.R. China
| | - Jianwei Xie
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P.R. China.,Department of Pediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangdong, P.R. China
| | - Xinyu Li
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangdong, P.R. China
| | - Jianpei Fang
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangdong, P.R. China
| |
Collapse
|
8
|
Li Y, Xie J, Li X, Fang J. Poly (ADP-ribosylation) of HMGB1 facilitates its acetylation and promotes HMGB1 translocation-associated chemotherapy-induced autophagy in leukaemia cells. Oncol Lett 2019; 19:368-378. [PMID: 31897149 PMCID: PMC6924101 DOI: 10.3892/ol.2019.11116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 07/26/2019] [Indexed: 12/15/2022] Open
Abstract
Acute lymphoblastic leukaemia (ALL) is one of the most common and curable types of cancer in paediatric patients. However, chemotherapeutic resistance is a difficult but common obstacle when treating leukaemia in the clinical setting. Studies have demonstrated that drug resistance is partly attributable to autophagy induced by multiple chemotherapeutic agents. As an evolutionarily conserved non-histone chromatin-binding protein, high mobility group box protein 1 (HMGB1) is considered to be an important factor in autophagy, and regulates autophagy at multiple levels via different subcellular localisations. In the present study, it was revealed that chemotherapeutic drugs induced autophagy in leukaemia cells and that translocation of HMGB1 from the nucleus to the cytoplasm is an important molecular event in this process. It was further demonstrated that poly (ADP-ribosylation) of HMGB1 facilitates its acetylation, thereby inducing HMGB1 translocation and ultimately promoting chemotherapy-induced autophagy in leukaemic cells. Targeted HMGB1 translocation may overcome chemotherapy-induced autophagy in leukaemia.
Collapse
Affiliation(s)
- Yunyao Li
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China.,Department of Paediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jianwei Xie
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China.,Department of Paediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Xinyu Li
- Department of Paediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jianpei Fang
- Department of Paediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
9
|
Yang S, Lee W, Lee BS, Lee C, Park EK, Ku SK, Bae JS. Aloin Reduces HMGB1-Mediated Septic Responses and Improves Survival in Septic Mice by Activation of the SIRT1 and PI3K/Nrf2/HO-1 Signaling Axis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:613-633. [PMID: 30966773 DOI: 10.1142/s0192415x19500320] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
High mobility group box 1 (HMGB1) is recognized as a late mediator of sepsis, and the inhibition of HMGB1 release and recovery of vascular barrier integrity have emerged as attractive therapeutic strategies for the management of sepsis. We tested the hypothesis that aloin induces sirtuin 1 (SIRT1) and heme oxygenase (HO)-1, which inhibit HMGB1 release in lipopolysaccharide (LPS)-stimulated cells, thereby inhibiting HMGB1-induced hyperpermeability and increasing the survival of septic mice. Aloin was administered after LPS or HMGB1 challenge, and the antiseptic activity of aloin was determined from measurements of permeability, activation of pro-inflammatory proteins and production of markers for tissue injury in HMGB1-activated human umbilical vein endothelial cells (HUVECs) and a cecal ligation and puncture (CLP)-induced sepsis mouse model. Aloin significantly reduced HMGB1 release in LPS-activated HUVECs via SIRT1-mediated HMGB1 deacetylation and the PI3K/Nrf2/heme oxygenase (HO)-1 signaling axis. Aloin also suppressed the production of tumor necrosis factor (TNF)- α and interleukin (IL)-6, as well as the activation of nuclear factor (NF)- κ B and extracellular signal-regulated kinase 1/2 (ERK 1/2) by HMGB1. Moreover, aloin restored HMGB1-mediated vascular disruption and inhibited vascular hyperpermeability in mice. In addition, treatment with aloin reduced the CLP-induced release of HMGB1, sepsis-related mortality and tissue injury in vivo. Our results suggest that aloin reduces HMGB1 release and sepsis-related mortality by activating SIRT1 and PI3K/Nrf2/HO-1 signals, indicating that aloin has potential for the treatment of sepsis.
Collapse
Affiliation(s)
- Sumin Yang
- * College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Wonhwa Lee
- * College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Bong-Seon Lee
- * College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Changhun Lee
- * College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Eui Kyun Park
- † Department of Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu 41940, Republic of Korea
| | - Sae-Kwang Ku
- ‡ Department of Histology and Anatomy, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Jong-Sup Bae
- * College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
10
|
Sun S, Adyshev D, Dudek S, Paul A, McColloch A, Cho M. Cholesterol-dependent Modulation of Stem Cell Biomechanics: Application to Adipogenesis. J Biomech Eng 2019; 141:2729412. [PMID: 30901381 DOI: 10.1115/1.4043253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Indexed: 11/08/2022]
Abstract
Cell mechanics has been shown to regulate stem cell differentiation. We have previously reported that altered cell stiffness of mesenchymal stem cells can delay or facilitate biochemically directed differentiation. One of the factors that can affect the cell stiffness is cholesterol. However, the effect of cholesterol on differentiation of human mesenchymal stem cells (hMSCs) remains elusive. In this paper, we demonstrate that cholesterol is involved in the modulation of the cell stiffness and subsequent adipogenic differentiation. Rapid cytoskeletal actin reorganization was evident and correlated with the cell's Young's modulus measured using atomic force microscopy (AFM). In addition, the level of membrane-bound cholesterol was found to increase during adipogenic differentiation and inversely varied with the cell stiffness. Furthermore, cholesterol played a key role in the regulation of the cell morphology and biomechanics, suggesting its crucial involvement in mechanotransduction. To better understand the underlying mechanisms, we investigated the effect of cholesterol on the membrane-cytoskeleton linker proteins (ezrin and moesin). Cholesterol depletion was found to up-regulate the ezrin expression which promoted cell spreading, increased Young's modulus, and hindered adipogenesis. In contrast, cholesterol enrichment increased the moesin expression, decreased Young's modulus, and induced cell rounding and facilitated adipogenesis. Taken together, cholesterol appears to regulate the stem cell mechanics and adipogenesis through the membrane-associated linker proteins.
Collapse
Affiliation(s)
- Shan Sun
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607
| | - Djanybek Adyshev
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607
| | - Steve Dudek
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607
| | - Amit Paul
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607
| | - Andrew McColloch
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019
| | - Michael Cho
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019
| |
Collapse
|
11
|
Chen M, Chen C, Yuan X, Chen X, Zheng F, Shao L, Zhang Z. Angiotensin II aggravates lipopolysaccharide induced human pulmonary microvascular endothelial cells permeability in high glucose status. Endocr J 2018; 65:717-725. [PMID: 29709898 DOI: 10.1507/endocrj.ej17-0477] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Lung infection is one of the most common infections in diabetes mellitus and is characterized by increased pulmonary microvascular endothelial permeability. Local Angiotensin II (AngII) plays an important role in the pathogenesis of lung diseases. However, whether AngII can aggravate diabetic infectious lung injury is not clear. Therefore, we investigated the effects of AngII on the permeability of human pulmonary microvascular endothelial cells (HPMVECs) challenged by lipopolysaccharide (LPS) in high glucose states in vitro. HPMVECs were divided into five groups: a control group (CON), a high glucose group (HG), an LPS + high glucose group (LH), an LPS + high glucose + AngII group (LHA), and an LPS + high glucose + Losartan group (LHL). The HPMVECs permeability as well as the F-actin levels, cytoskeleton, apoptosis and TNF-α concentrations were evaluated. Compared to the CON group, the HG, LH and LHA groups had significantly higher cellular permeability, cellular apoptosis and TNF-α levels, with more extensive cytoskeletal damage and lower F-actin levels. Additionally, cells in the LHA group exhibited significantly elevated permeability, apoptosis and TNF-α concentrations, lower F-actin levels and more extensive cytoskeletal damage than either the LH or HG group. However, compared to the HG or LH group, the LHL group showed significantly lower cellular permeability, cell apoptosis, cytoskeletal damage and TNF-α concentrations and higher F-actin levels. This study suggests that in a diabetic infectious lung injury cellular model, AngII could aggravate the permeability of HPMVEC via F-actin dynamics and cell apoptosis. Furthermore, blocking the Angiotension II Type 1 Receptor could significantly alleviate the hyperpermeability of HPMVECs.
Collapse
Affiliation(s)
- Min Chen
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Hubei 430071, China
| | - Chang Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei 430071, China
| | - Xiaohui Yuan
- Department of Anesthesiology, Tongren Hospital of Wuhan University & Wuhan Third Hospital, Hubei 430022, China
| | - Xiaoqi Chen
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Hubei 430071, China
| | - Feng Zheng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei 430071, China
| | - Liang Shao
- Department of Hematology, Zhongnan Hospital of Wuhan University, Hubei 430071, China
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei 430071, China
| |
Collapse
|
12
|
Lee Y, Lee W, Chang H, Kim S, Kim J, Bae J. Testican‐1, as a novel diagnosis of sepsis. J Cell Biochem 2018; 119:4216-4223. [DOI: 10.1002/jcb.26661] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/04/2018] [Indexed: 11/11/2022]
Affiliation(s)
- Yuri Lee
- College of PharmacyCMRIResearch Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi‐Omics based Creative Drug Research TeamKyungpook National UniversityDaeguRepublic of Korea
| | - Wonhwa Lee
- College of PharmacyCMRIResearch Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi‐Omics based Creative Drug Research TeamKyungpook National UniversityDaeguRepublic of Korea
- Aging Research CenterKorea Research Institute of Bioscience and BiotechnologyDeajeonRepublic of Korea
| | - Hyun‐Ha Chang
- Department of Internal MedicineSchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
| | - Shin‐Woo Kim
- Department of Internal MedicineSchool of MedicineKyungpook National UniversityDaeguRepublic of Korea
| | - Jaehong Kim
- Department of BiochemistrySchool of MedicineGachon UniversityIncheonRepublic of Korea
| | - Jong‐Sup Bae
- College of PharmacyCMRIResearch Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi‐Omics based Creative Drug Research TeamKyungpook National UniversityDaeguRepublic of Korea
| |
Collapse
|
13
|
Sung E, Kwon OK, Lee JM, Lee S. Proteomics approach to identify novel metastatic bone markers from the secretome of PC-3 prostate cancer cells. Electrophoresis 2017. [PMID: 28627741 DOI: 10.1002/elps.201700052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Prostate cancer is the leading type of cancer diagnosed, and the most frequent cause of worldwide male cancer-related deaths annually. The limitations of current prostate cancer screening tests demand the identification of novel biomarkers for the early diagnosis of prostate cancer bone metastasis. In the present study, we performed a proteomic analysis of secreted proteins from the prostate cancer bone metastasis cell line, PC-3, and the normal prostate cell line, RWPE-1. We thus quantified 917 proteins, of which 68 were found to be secreted at higher levels by PC-3 than by RWPE-1 cells via LC-MS/MS. To characterize the highly secreted proteins in the PC-3 cell line and thereby identify biomarker proteins, we divided the quantifiable proteins into four quantitative categories (Q1-Q4). The KEGG lysine degradation and osteoclast differentiation pathways were demonstrated to be enriched in the highly secreted Q4 protein group. Transforming growth factor (TGF) beta family proteins related to osteoclast differentiation were identified as key regulators of PC-3 cell proliferation. Immunoblotting was used to confirm the observed high level of pentraxin, follistatin, TGF-beta family members, and serpin B3 secretion by PC-3 cells. From the collective results of the present study, we suggest that serpin B3 is a promising novel biomarker candidate for the diagnosis of prostate cancer bone metastasis.
Collapse
Affiliation(s)
- EunJi Sung
- BK21 Plus KNU Multi-Omics-based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Oh Kwang Kwon
- BK21 Plus KNU Multi-Omics-based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Jae-Mok Lee
- Department of Periodontology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Sangkyu Lee
- BK21 Plus KNU Multi-Omics-based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
14
|
Deng H, Wang C, Chang DY, Hu N, Chen M, Zhao MH. High mobility group box-1 contributes to anti-myeloperoxidase antibody-induced glomerular endothelial cell injury through a moesin-dependent route. Arthritis Res Ther 2017; 19:125. [PMID: 28587670 PMCID: PMC5461689 DOI: 10.1186/s13075-017-1339-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 05/18/2017] [Indexed: 02/06/2023] Open
Abstract
Background Our previous study found that circulating and urinary levels of high mobility group box-1 (HMGB1) were closely associated with disease activity in patients with antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). Moreover, HMGB1 participates in ANCA-induced neutrophil activation. Cross-reactivity between moesin and anti-myeloperoxidase (MPO) antibody has been reported in both human and mouse. The current study investigated whether HMGB1 participated in MPO-ANCA-induced glomerular endothelial cell (GEnC) injury, which is one of the most important aspects in the pathogenesis of AAV. Methods The effects of HMGB1 on expression of moesin on GEnCs and anti-MPO antibody binding to GEnCs were measured. MPO expression on GEnCs was explored. The effects of HMGB1 in MPO-ANCA induced GEnC injury were measured, during which the role of moesin was explored. Antagonists for various relevant receptors were employed. Results Sera from AAV patients at the active stage could mediate GEnC injury, while this effect could be attenuated by preblocking HMGB1. HMGB1 could increase the expression of moesin on GEnCs and the binding of anti-MPO antibody to moesin. The colocalization of moesin expression and anti-MPO antibody binding can be detected. Little, if any, MPO was expressed in GEnCs. HMGB1 increased GEnC activation and injury in the presence of patient-derived MPO-ANCA-positive IgGs through moesin. The effects of HMGB1 on expression of moesin on GEnCs, anti-MPO antibody binding to GEnCs, GEnC activation and injury were mainly toll like receptor 4 (TLR4) dependent. Conclusions HMGB1 can increase the expression of moesin but not MPO on GEnCs, and can further participate in MPO-ANCA-induced GEnC activation and injury by cross-reactivity between moesin and anti-MPO antibody. Electronic supplementary material The online version of this article (doi:10.1186/s13075-017-1339-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hui Deng
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China
| | - Chen Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China
| | - Dong-Yuan Chang
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China
| | - Nan Hu
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China
| | - Min Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.
| | - Ming-Hui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China
| |
Collapse
|
15
|
Schnoor M, García Ponce A, Vadillo E, Pelayo R, Rossaint J, Zarbock A. Actin dynamics in the regulation of endothelial barrier functions and neutrophil recruitment during endotoxemia and sepsis. Cell Mol Life Sci 2017; 74:1985-1997. [PMID: 28154894 PMCID: PMC11107778 DOI: 10.1007/s00018-016-2449-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 01/20/2023]
Abstract
Sepsis is a leading cause of death worldwide. Increased vascular permeability is a major hallmark of sepsis. Dynamic alterations in actin fiber formation play an important role in the regulation of endothelial barrier functions and thus vascular permeability. Endothelial integrity requires a delicate balance between the formation of cortical actin filaments that maintain endothelial cell contact stability and the formation of actin stress fibers that generate pulling forces, and thus compromise endothelial cell contact stability. Current research has revealed multiple molecular pathways that regulate actin dynamics and endothelial barrier dysfunction during sepsis. These include intracellular signaling proteins of the small GTPases family (e.g., Rap1, RhoA and Rac1) as well as the molecules that are directly acting on the actomyosin cytoskeleton such as myosin light chain kinase and Rho kinases. Another hallmark of sepsis is an excessive recruitment of neutrophils that also involves changes in the actin cytoskeleton in both endothelial cells and neutrophils. This review focuses on the available evidence about molecules that control actin dynamics and regulate endothelial barrier functions and neutrophil recruitment. We also discuss treatment strategies using pharmaceutical enzyme inhibitors to target excessive vascular permeability and leukocyte recruitment in septic patients.
Collapse
Affiliation(s)
- Michael Schnoor
- Department for Molecular Biomedicine, Centre for Investigation and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. IPN 2508, San Pedro Zacatenco, GAM, 07360, Mexico City, Mexico.
| | - Alexander García Ponce
- Department for Molecular Biomedicine, Centre for Investigation and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. IPN 2508, San Pedro Zacatenco, GAM, 07360, Mexico City, Mexico
| | - Eduardo Vadillo
- Department for Molecular Biomedicine, Centre for Investigation and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. IPN 2508, San Pedro Zacatenco, GAM, 07360, Mexico City, Mexico
| | - Rosana Pelayo
- Oncology Research Unit, National Medical Center, Mexican Institute for Social Security, 06720, Mexico City, Mexico
| | - Jan Rossaint
- Department of Anaesthesiology, Critical Care and Pain Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Alexander Zarbock
- Department of Anaesthesiology, Critical Care and Pain Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany.
| |
Collapse
|
16
|
RAGE deficiency attenuates the protective effect of Lidocaine against sepsis-induced acute lung injury. Inflammation 2017; 40:601-611. [DOI: 10.1007/s10753-016-0507-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
17
|
Wang C, Chang DY, Chen M, Zhao MH. HMGB1 contributes to glomerular endothelial cell injury in ANCA-associated vasculitis through enhancing endothelium-neutrophil interactions. J Cell Mol Med 2017; 21:1351-1360. [PMID: 28181422 PMCID: PMC5487910 DOI: 10.1111/jcmm.13065] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 11/17/2016] [Indexed: 12/19/2022] Open
Abstract
Our previous studies demonstrated that high mobility group box‐1 (HMGB1), a typical damage‐associated molecular pattern (DAMP) protein, is associated with the disease activity of antineutrophil cytoplasmic antibody (ANCA)‐associated vasculitis (AAV). Moreover, HMGB1 participates in ANCA‐induced neutrophil activation. The current study aimed to investigate whether HMGB1 regulated the interaction between neutrophils and glomerular endothelial cells (GEnC) in the presence of ANCA. Correlation analysis on HMGB1 levels in AAV patients and soluble intercellular cell adhesion molecule‐1 (sICAM‐1) levels or vascular endothelial growth factor (VEGF) levels, which are markers of endothelial cell activation, was performed. The effect of HMGB1 on neutrophil migration towards GEnC, respiratory burst and degranulation of neutrophils in coculture conditions with GEnC was measured. The activation of neutrophils, the activation and injury of GEnC, and the consequent pathogenic role of injured GEnC were evaluated. Plasma levels of HMGB1 correlated with sICAM‐1 and VEGF (r = 0.73, P < 0.01; r = 0.41, P = 0.04) in AAV patients. HMGB1 increased neutrophil migration towards GEnC, as well as respiratory burst and degranulation of neutrophils in the presence of ANCA in the coculture system. In the presence of robust neutrophil activation, GEnC were further activated and injured in the coculture system of GEnC and neutrophils. In addition, injured GEnC could produce TF‐positive leuco‐endothelial microparticles and endothelin‐1 (ET‐1), while NF‐κB was phosphorylated (S529) in the injured GEnC. Plasma levels of HMGB1 correlated with endothelial cell activation in AAV patients. HMGB1 amplified neutrophil activation and the activation and injury of GEnC in the presence of ANCA.
Collapse
Affiliation(s)
- Chen Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Dong-Yuan Chang
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Min Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Ming-Hui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| |
Collapse
|
18
|
Singh A, Agarwal S, Doreswamy SM, Chakkarapani AA, Murthy P, Kajale NA, Mughal Z, Khadilkar V, Chiplonkar SA, Khadilkar A, Ma J, Lu H. Psychosocial care and its association with severe acute malnutrition. Indian Pediatr 2016; 53:431-2. [DOI: 10.1007/s13312-016-0870-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
19
|
Posthemorrhagic shock mesenteric lymph enhances monolayer permeability via F-actin and VE-cadherin. J Surg Res 2016; 203:47-55. [PMID: 27338534 DOI: 10.1016/j.jss.2016.01.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 01/05/2016] [Accepted: 01/27/2016] [Indexed: 11/22/2022]
Abstract
BACKGROUND Vascular hyperpermeability plays a critical role in the development of refractory hypotension after severe hemorrhagic shock. Posthemorrhagic shock mesenteric lymph (PHSML) return has been shown to be involved in regulation of vascular hyperpermeability. The present study was conducted to investigate the effect of PHSML on permeability of endothelial cells in vitro. MATERIALS AND METHODS A hemorrhagic shock model (40 ± 2 mm Hg for 90 min, followed by fluid resuscitation) was used for collection of PHSML. Two separated PHSMLs were collected from period 0-3 h (early) and period 3-6 h (late) after resuscitation and diluted into concentration of 4% or 10%. The human umbilical vein endothelial cells (HUVECs) were then treated with these PHSMLs for 6 h. The monolayer cellular permeability to FITC-albumin was observed by using the costar transwell system. The multiple approaches including scanning electron microscope, fluorescent cytochemistry staining, and Western blotting were also used to assess the changes in cellular morphologic and the expressions of F-actin and VE-cadherin. RESULTS The treatments with either early or late PHSML resulted in morphologic injuries, increased cellular permeability, and decreased expression of F-actin in HUVECs. In contrast, only early PHSML, but not late PHSML, reduced the VE-cadherin expression. CONCLUSIONS These results indicate that the PHSML in vitro increases the cellular permeability of HUVECs through suppression of F-actin and VE-cadherin.
Collapse
|
20
|
Wang C, de Souza AW, Westra J, Bijl M, Chen M, Zhao MH, Kallenberg CG. Emerging role of high mobility group box 1 in ANCA-associated vasculitis. Autoimmun Rev 2015. [DOI: 10.1016/j.autrev.2015.07.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|