1
|
Prideaux EB, Boyle DL, Choi E, Buckner JH, Robinson WH, Holers VM, Deane KD, Firestein GS, Wang W. Epigenetic trajectory predicts development of clinical rheumatoid arthritis in ACPA+ individuals: Targeting Immune Responses for Prevention of Rheumatoid Arthritis (TIP-RA). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.15.618490. [PMID: 39463978 PMCID: PMC11507690 DOI: 10.1101/2024.10.15.618490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
OBJECTIVE The presence of autoantibodies to citrullinated protein antigens (ACPAs) in the absence of clinically-apparent inflammatory arthritis (IA) identifies individuals at-risk for developing future clinical rheumatoid arthritis (RA). However, it is unclear why some ACPA+ individuals convert to clinical RA while others do not. We explored the possibility in the Targeting Immune Responses for Prevention of Rheumatoid Arthritis (TIP-RA) study that epigenetic remodeling is part of the trajectory from an at-risk state to clinical disease and identifies novel biomarkers associated with conversion to clinical RA. METHODS ACPA- Controls, ACPA+ At-Risk, and Early RA individuals were followed for up to 5 years, including obtaining blood samples annually and at RA diagnosis. Peripheral blood mononuclear cells (PBMCs) were separated into CD19+ B cells, memory CD4+ T cells, and naive CD4+ T cells using antibodies and magnetic beads. Genome-wide methylation within each cell lineage was assayed using the Illumina MethylationEPIC v1.0 beadchip. ACPA+ At-Risk participants who did or did not develop RA were designated Pre-RA or Non-converters, respectively. Differentially methylated loci (DML) were selected using the Limma software package. Using the Caret package, we constructed machine learning models in test and validation cohorts and identified the most predictive loci of clinical RA conversion. RESULTS Cross-sectional differential methylation analysis at baseline revealed DMLs that distinguish the Pre-RA methylome from ACPA+ Non-converters, the latter which closely resembled ACPA- Controls. Genes overlapping these DMLs correspond to aberrant NOTCH signaling and DNA repair pathways in B cells. Longitudinal analysis showed that ACPA- Control and ACPA+ Non-converter methylomes are relatively constant. In contrast, the Pre-RA methylome remodeled along a dynamic RA methylome trajectory characterized by epigenetic changes in active regulatory elements. Clinical conversion to RA, defined based on diagnosis, marked an epigenetic inflection point for cell cycle pathways in B cells and adaptive immunity pathways in naive T cells. Machine learning revealed individual loci associated with RA conversion. This model significantly outperformed autoantibodies plus acute phase reactants as predictors of RA conversion. CONCLUSION DNA methylation is a dynamic process in ACPA+ individuals at-risk for developing RA that eventually transition to clinical disease. In contrast, non-converters and controls have stable methylomes. The accumulation of epigenetic marks over time prior to conversion to clinical RA conforms to pathways that are associated with immunity and can be used to identify potential pathogenic pathways for therapeutic targeting and/or use as prognostic biomarkers.
Collapse
|
2
|
Neves RL, Marem A, Carmona B, Arata JG, Cyrillo Ramos MP, Justo GZ, Machado de Melo FH, Oliveira V, Icimoto MY. Expression of thimet oligopeptidase (THOP) modulated by oxidative stress in human multidrug resistant (MDR) leukemia cells. Biochimie 2023; 212:21-30. [PMID: 36997147 DOI: 10.1016/j.biochi.2023.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/03/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023]
Abstract
Thimet oligopeptidase (THOP) is a cytosolic metallopeptidase known to regulate the fate of post-proteasomal peptides, protein turnover and peptide selection in the antigen presentation machinery (APM) system. Oxidative stress influences THOP expression and regulates its proteolytic activity, generating variable cytosolic peptide levels, possibly affecting the immune evasion of tumor cells. In the present work, we examined the association between THOP expression/activity and stress oxidative resistance in human leukemia cells using the K562 cell line, a chronic myeloid leukemia (CML), and the multidrug-resistant (MDR) Lucena 1 (K562-derived MDR cell line) as model. The Lucena 1 phenotype was validated under vincristine treatment and the relative THOP1 mRNA levels and protein expression compared to K562 cell line. Our data demonstrated increased THOP1 gene and protein levels in K562 cells in contrast to the oxidative-resistant Lucena 1, even after H2O2 treatment, suggesting an oxidative stress dependence in THOP regulation. Further, it was observed higher basal levels of reactive oxygen species (ROS) in K562 compared to Lucena 1 cell line using DHE fluorescent probe. Since THOP activity is dependent on its oligomeric state, we also compared its proteolytic activity under reducing agent treatment, which demonstrated that its function modulation with respect to changes in redox state. Finally, the mRNA expression and FACS analyses demonstrated a reduced expression of MHC I only in K562 cell line. In conclusion, our results highlight THOP redox modulation, which could influence antigen presentation in multidrug resistant leukemia cells.
Collapse
Affiliation(s)
- Raquel Leão Neves
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Alyne Marem
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Bruno Carmona
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Júlia Galanakis Arata
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | | | - Giselle Zenker Justo
- Departamento de Bioquímica, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil; Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo, Diadema, São Paulo, Brazil
| | | | - Vitor Oliveira
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil.
| | - Marcelo Yudi Icimoto
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil; Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, Oregon Health and Science University, Portland, OR, United States.
| |
Collapse
|
3
|
Liu Y, Sigman J, Bruce L, Wolfson A. Thimet Oligopeptidase—A Classical Enzyme with New Function and New Form. IMMUNO 2021; 1:332-346. [DOI: 10.3390/immuno1040022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Peptidases generate bioactive peptides that can regulate cell signaling and mediate intercellular communication. While the processing of peptide precursors is initiated intracellularly, some modifications by peptidases may be conducted extracellularly. Thimet oligopeptidase (TOP) is a peptidase that processes neuroendocrine peptides with roles in mood, metabolism, and immune responses, among other functions. TOP also hydrolyzes angiotensin I to angiotensin 1–7, which may be involved in the pathophysiology of COVID-19 infection. Although TOP is primarily cytosolic, it can also be associated with the cell plasma membrane or secreted to the extracellular space. Recent work indicates that membrane-associated TOP can be released with extracellular vesicles (EVs) to the extracellular space. Here we briefly summarize the enzyme’s classical function in extracellular processing of neuroendocrine peptides, as well as its more recently understood role in intracellular processing of various peptides that impact human diseases. Finally, we discuss new findings of EV-associated TOP in the extracellular space.
Collapse
Affiliation(s)
- Yu Liu
- Department of Chemistry, Wellesley College, Wellesley, MA 02481, USA
- Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
- Department of GI/Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey Sigman
- Department of Chemistry, St. Mary’s College of California, Moraga, CA 94575, USA
| | - Lisa Bruce
- Department of Chemistry, Wellesley College, Wellesley, MA 02481, USA
- Invetx, Boston, MA 02108, USA
| | - Adele Wolfson
- Department of Chemistry, Wellesley College, Wellesley, MA 02481, USA
| |
Collapse
|
4
|
Thimet Oligopeptidase Biochemical and Biological Significances: Past, Present, and Future Directions. Biomolecules 2020; 10:biom10091229. [PMID: 32847123 PMCID: PMC7565970 DOI: 10.3390/biom10091229] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/15/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
Thimet oligopeptidase (EC 3.4.24.15; EP24.15, THOP1) is a metallopeptidase ubiquitously distributed in mammalian tissues. Beyond its previously well characterized role in major histocompatibility class I (MHC-I) antigen presentation, the recent characterization of the THOP1 C57BL6/N null mice (THOP1−/−) phenotype suggests new key functions for THOP1 in hyperlipidic diet-induced obesity, insulin resistance and non-alcoholic liver steatosis. Distinctive levels of specific intracellular peptides (InPeps), genes and microRNAs were observed when comparing wild type C57BL6/N to THOP1−/− fed either standard or hyperlipidic diets. A possible novel mechanism of action was suggested for InPeps processed by THOP1, which could be modulating protein-protein interactions and microRNA processing, thus affecting the phenotype. Together, research into the biochemical and biomedical significance of THOP1 suggests that degradation by the proteasome is a step in the processing of various proteins, not merely for ending their existence. This allows many functional peptides to be generated by proteasomal degradation in order to, for example, control mRNA translation and the formation of protein complexes.
Collapse
|
5
|
Rochfort KD, Carroll LS, Barabas P, Curtis TM, Ambati BK, Barron N, Cummins PM. COMP-Ang1 Stabilizes Hyperglycemic Disruption of Blood-Retinal Barrier Phenotype in Human Retinal Microvascular Endothelial Cells. Invest Ophthalmol Vis Sci 2019; 60:3547-3555. [PMID: 31415078 PMCID: PMC6699794 DOI: 10.1167/iovs.19-27644] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Purpose Current treatments for diabetic retinopathy (DR) have considerable limitations, underpinning the need for new therapeutic options. In this article, the ability of an engineered angiopoietin-1 variant (COMP-Ang1) to ameliorate the injurious effects of hyperglycemia on barrier integrity in a human retinal microvascular endothelial cell (HRMvEC) model is comprehensively investigated. Methods Confluent HRMvECs were treated (0–72 hours) with d-glucose (5 or 30 mM) in the absence and presence of COMP-Ang1 (10–200 ng/mL). l-glucose (30 mM) was used as osmotic control. Posttreatment, intact cell monolayers were monitored for permeability to FITC-dextran 40 kDa. Cells were also harvested for analysis of interendothelial junction targets by RT-qPCR and Western blotting. The impact of receptor tyrosine kinase Tie2 gene silencing on COMP-Ang1 efficacy was also evaluated. Results Treatment with 30 mM d-glucose (but not l-glucose) demonstrated a time-dependent elevation in the mean rate of FITC-dextran diffusion across intact HRMvEC monolayers, in parallel with significant reductions in mRNA/protein levels of occludin, claudin-5, ZO-1, and VE-Cadherin. These effects were all attenuated by COMP-Ang1 in a concentration-dependent fashion, with 200 ng/mL recovering barrier function by ∼88%, and recovering reduced interendothelial junction protein levels by more than 50%. Finally, Tie2 knockdown by small interfering RNA silencing blocked the ability of COMP-Ang1 to mitigate against hyperglycemia-induced permeabilization of HRMvECs and depletion of junctional expression levels. Conclusions In summary, this article presents a reproducible in vitro cell study that quantifies the concentration-dependent efficacy of COMP-Ang1 to mitigate the injurious effects of hyperglycemic challenge on HRMvEC barrier properties via Tie2-mediated signaling.
Collapse
Affiliation(s)
- Keith D Rochfort
- School of Biotechnology, Dublin City University, Dublin, Ireland.,National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Lara S Carroll
- John A. Moran Eye Center, University of Utah, Salt Lake City, Utah, United States
| | - Peter Barabas
- Wellcome-Wolfson Institute for Experimental Medicine, Queens' University Belfast, Northern Ireland, United Kingdom
| | - Timothy M Curtis
- Wellcome-Wolfson Institute for Experimental Medicine, Queens' University Belfast, Northern Ireland, United Kingdom
| | - Balamurali K Ambati
- John A. Moran Eye Center, University of Utah, Salt Lake City, Utah, United States
| | - Niall Barron
- National Institute for Bioprocessing Research & Training (NIBRT), Dublin, Ireland
| | - Philip M Cummins
- School of Biotechnology, Dublin City University, Dublin, Ireland.,National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| |
Collapse
|
6
|
Li J, Zhao Y, Coleman P, Chen J, Ting KK, Choi JP, Zheng X, Vadas MA, Gamble JR. Low fluid shear stress conditions contribute to activation of cerebral cavernous malformation signalling pathways. Biochim Biophys Acta Mol Basis Dis 2019; 1865:165519. [DOI: 10.1016/j.bbadis.2019.07.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 06/18/2019] [Accepted: 07/27/2019] [Indexed: 02/07/2023]
|
7
|
Hashimoto S, Onoe T, Banshodani M, Taguchi K, Tanaka Y, Ohdan H. Postoperative Portal Hypertension Enhances Alloimmune Responses after Living-Donor Liver Transplantation in Patients and in a Mouse Model. THE JOURNAL OF IMMUNOLOGY 2019; 203:1392-1403. [PMID: 31331971 DOI: 10.4049/jimmunol.1701147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 06/25/2019] [Indexed: 12/29/2022]
Abstract
Controlling portal vein pressure in living-donor liver transplantation has received increased attention owing to its potential importance for graft survival. Portal hypertension may lead to the activation of liver-resident APCs, including liver sinusoidal endothelial cells (LSECs), which have immunological tolerogenic capacity. We investigated the effects of portal hypertension on graft survival and the antidonor immune response using clinical data and a mouse model. We categorized patients (n = 136) according to their portal vein pressure values at the end of surgery. Using propensity score-matching analyses, we found that portal hypertension was significantly associated with a higher antidonor immune response and incidence of acute rejection. To investigate the mechanism, we performed an allogeneic coculture assay using a 70% hepatectomized (HTx) mouse model with or without a portosystemic shunt. Liver cells from HTx mice without a shunt exhibited a significantly greater anti-BALB/c B6 T cell response than those from sham-operated mice or HTx mice with a shunt. LSECs from sham-operated mice, but not from HTx mice, suppressed the B6 T cell alloresponse in a dose-dependent manner. Furthermore, LSECs from HTx mice without a shunt showed significantly downregulated MHC class I/II and programmed death-ligand 1 expression, and those from mice with a shunt showed recovered expression of these molecules. Postoperative portal hypertension enhances alloimmune responses in recipients after living-donor liver transplantation, likely due, in part, to the impaired immune-suppression capacity of LSECs.
Collapse
Affiliation(s)
- Shinji Hashimoto
- Department of Gastroenterological and Transplant Surgery, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-0037, Japan; and
| | - Takashi Onoe
- Department of Gastroenterological and Transplant Surgery, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-0037, Japan; and .,Institute for Clinical Research, National Hospital Organization, Kure Medical Center/Chugoku Cancer Center, Kure 737-0023, Japan
| | - Masataka Banshodani
- Department of Gastroenterological and Transplant Surgery, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-0037, Japan; and
| | - Kazuhiro Taguchi
- Department of Gastroenterological and Transplant Surgery, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-0037, Japan; and.,Institute for Clinical Research, National Hospital Organization, Kure Medical Center/Chugoku Cancer Center, Kure 737-0023, Japan
| | - Yuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-0037, Japan; and
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-0037, Japan; and
| |
Collapse
|
8
|
Rochfort KD, Collins LE, McLoughlin A, Cummins PM. Tumour necrosis factor-α-mediated disruption of cerebrovascular endothelial barrier integrity in vitro involves the production of proinflammatory interleukin-6. J Neurochem 2015; 136:564-72. [PMID: 26499872 DOI: 10.1111/jnc.13408] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 10/02/2015] [Accepted: 10/12/2015] [Indexed: 12/12/2022]
Abstract
The co-involvement of tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6) during blood-brain barrier (BBB) injury has been reported in various models of neuroinflammation, although the precise functional interplay between these archetypal proinflammatory cytokines remains largely undefined within this context. In the current paper, we tested the hypothesis that TNF-α-mediated BBB disruption is measurably attributable in-part to induction of microvascular endothelial IL-6 production. In initial experiments, we observed that treatment of human brain microvascular endothelial cells (HBMvECs) with TNF-α (0-100 ng/mL, 0-24 h) robustly elicited both time- and dose-dependent induction of IL-6 expression and release, as well as expression of the IL-6 family receptor, GP130. Further experiments demonstrated that the TNF-α-dependent generation of reactive oxygen species, down-regulation of adherens/tight junction proteins, and concomitant elevation of HBMvEC permeability, were all significantly attenuated by blockade of IL-6 signalling using either an anti-IL-6 neutralizing antibody or an IL-6 siRNA. Based on these observations, we conclude that TNF-α treatment of HBMvECs in vitro activates IL-6 production and signalling, events that were shown to synergize with TNF-α actions to elicit HBMvEC permeabilization. These novel findings offer a constructive insight into the specific contribution of downstream cytokine induction to the injurious actions of TNF-α at the BBB microvascular endothelium interface. The co-involvement of tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6) during blood-brain barrier (BBB) injury has been widely reported. Using human brain microvascular endothelial cells (HBMvEC), we show that TNF-α-mediated BBB disruption is measurably attributable in-part to induction of endothelial IL-6 production and signalling. We demonstrate that the TNF-α-dependent generation of reactive oxygen species (ROS), down-regulation of interendothelial junctions, and concomitant elevation of HBMvEC permeability, could be significantly attenuated by using either an IL-6 neutralizing antibody or an IL-6-specific siRNA. These findings provide insight into the complex nature of proinflammatory cytokine injury at the BBB microvascular endothelium interface.
Collapse
Affiliation(s)
- Keith D Rochfort
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Laura E Collins
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Alisha McLoughlin
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Philip M Cummins
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland.,Centre for Preventive Medicine, Dublin City University, Glasnevin, Dublin, Ireland
| |
Collapse
|
9
|
Shear-dependent attenuation of cellular ROS levels can suppress proinflammatory cytokine injury to human brain microvascular endothelial barrier properties. J Cereb Blood Flow Metab 2015; 35:1648-56. [PMID: 25991069 PMCID: PMC4640321 DOI: 10.1038/jcbfm.2015.102] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/22/2015] [Indexed: 12/21/2022]
Abstract
The regulatory interplay between laminar shear stress and proinflammatory cytokines during homeostatic maintenance of the brain microvascular endothelium is largely undefined. We hypothesized that laminar shear could counteract the injurious actions of proinflammatory cytokines on human brain microvascular endothelial cell (HBMvEC) barrier properties, in-part through suppression of cellular redox signaling. For these investigations, HBMvECs were exposed to either shear stress (8 dynes/cm(2), 24 hours) or cytokines (tumor necrosis factor-α (TNF-α) or interleukin-6 (IL-6), 0 to 100 ng/mL, 6 or 18 hours). Human brain microvascular endothelial cell 'preshearing'±cytokine exposure was also performed. Either cytokine dose-dependently decreased expression and increased phosphorylation (pTyr/pThr) of interendothelial occludin, claudin-5, and vascular endothelial-cadherin; observations directly correlating to endothelial barrier reduction, and in precise contrast to effects seen with shear. We further observed that, relative to unsheared cells, HBMvECs presheared for 24 hours exhibited significantly reduced reactive oxygen species production and barrier permeabilization in response to either TNF-α or IL-6 treatment. Shear also downregulated NADPH oxidase (nicotinamide adenine dinucleotide phosphate-oxidase) activation in HBMvECs, as manifested in the reduced expression and coassociation of gp91phox and p47phox. These findings lead us to conclude that physiologic shear can protect the brain microvascular endothelium from injurious cytokine effects on interendothelial junctions and barrier function by regulating the cellular redox state in-part through NADPH oxidase inhibition.
Collapse
|
10
|
Cytokine-mediated dysregulation of zonula occludens-1 properties in human brain microvascular endothelium. Microvasc Res 2015; 100:48-53. [DOI: 10.1016/j.mvr.2015.04.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 04/29/2015] [Accepted: 04/29/2015] [Indexed: 11/21/2022]
|
11
|
Rochfort KD, Cummins PM. Thrombomodulin regulation in human brain microvascular endothelial cells in vitro: role of cytokines and shear stress. Microvasc Res 2014; 97:1-5. [PMID: 25250518 DOI: 10.1016/j.mvr.2014.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 08/23/2014] [Accepted: 09/13/2014] [Indexed: 11/18/2022]
Abstract
Thrombomodulin (TM), an important determinant of blood vessel homeostasis, is expressed on the luminal vascular endothelial cell surface and is released into serum in response to circulatory signals. This includes the cerebrovascular endothelium, where the anti-coagulant and anti-inflammatory properties of TM are thought to be critical to the brain microcirculation and blood-brain barrier (BBB) integrity. Much is still unknown however about how circulatory stimuli may regulate TM activity within the brain microvasculature. To address this, the current short paper investigated the effects of opposing regulatory signals, namely cytokines (TNF-α, IL-6) and laminar shear stress, on the cellular levels and release of TM in cultured human brain microvascular endothelial cells (HBMvECs). Treatment of confluent HBMvECs with either TNF-α or IL-6 (100ng/ml, 18h) reduced TM protein levels by up to 70%, whilst inducing TM release into media by up to 4.4 and 5.5 fold, respectively. The effects of either cytokine (0-100ng/ml) on TM protein levels (6 or 18h) and release (0-18h) were also found to be concentration- and time-dependent. Either cytokine (100ng/ml, 24-72h) also reduced TM mRNA levels by >50%. When exposed to laminar shear stress for 24h at 8dyn/cm(2) (SI unit equivalent=0.8Pa), TM protein levels were upregulated by 65% in parallel with a 2-fold increase in TM mRNA levels. Shear stress also proved to be a much more potent stimulus for TM release from HBMvECs, yielding media TM levels of 1000pg/10(5) cells, when compared to 175 and 210pg/10(5) cells for TNF-α and IL-6, respectively, after parallel 18h treatments. Finally, shear-conditioned media was found to completely block thrombin-induced permeabilization of HBMvECs, confirming the functional efficacy of released TM. In summary, our data indicate that TM is differentially regulated within cultured HBMvECs by humoral and biomechanical signals.
Collapse
Affiliation(s)
- Keith D Rochfort
- School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland; Centre for Preventive Medicine, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Philip M Cummins
- School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland; Centre for Preventive Medicine, Dublin City University, Glasnevin, Dublin 9, Ireland.
| |
Collapse
|
12
|
Martin FA, McLoughlin A, Rochfort KD, Davenport C, Murphy RP, Cummins PM. Regulation of thrombomodulin expression and release in human aortic endothelial cells by cyclic strain. PLoS One 2014; 9:e108254. [PMID: 25238231 PMCID: PMC4169621 DOI: 10.1371/journal.pone.0108254] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 08/19/2014] [Indexed: 01/10/2023] Open
Abstract
Background and Objectives Thrombomodulin (TM), an integral membrane glycoprotein expressed on the lumenal surface of vascular endothelial cells, promotes anti-coagulant and anti-inflammatory properties. Release of functional TM from the endothelium surface into plasma has also been reported. Much is still unknown however about how endothelial TM is regulated by physiologic hemodynamic forces (and particularly cyclic strain) intrinsic to endothelial-mediated vascular homeostasis. Methods This study employed human aortic endothelial cells (HAECs) to investigate the effects of equibiaxial cyclic strain (7.5%, 60 cycles/min, 24 hrs), and to a lesser extent, laminar shear stress (10 dynes/cm2, 24 hrs), on TM expression and release. Time-, dose- and frequency-dependency studies were performed. Results Our initial studies demonstrated that cyclic strain strongly downregulated TM expression in a p38- and receptor tyrosine kinase-dependent manner. This was in contrast to the upregulatory effect of shear stress. Moreover, both forces significantly upregulated TM release over a 48 hr period. With continuing focus on the cyclic strain-induced TM release, we noted both dose (0–7.5%) and frequency (0.5–2.0 Hz) dependency, with no attenuation of strain-induced TM release observed following inhibition of MAP kinases (p38, ERK-1/2), receptor tyrosine kinase, or eNOS. The concerted impact of cyclic strain and inflammatory mediators on TM release from HAECs was also investigated. In this respect, both TNFα (100 ng/ml) and ox-LDL (10–50 µg/ml) appeared to potentiate strain-induced TM release. Finally, inhibition of neither MMPs (GM6001) nor rhomboids (3,4-dichloroisocoumarin) had any effect on strain-induced TM release. However, significantly elevated levels (2.1 fold) of TM were observed in isolated microparticle fractions following 7.5% strain for 24 hrs. Conclusions A preliminary in vitro investigation into the effects of cyclic strain on TM in HAECs is presented. Physiologic cyclic strain was observed to downregulate TM expression, whilst upregulating in a time-, dose- and frequency-dependent manner the release of TM.
Collapse
Affiliation(s)
- Fiona A. Martin
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Alisha McLoughlin
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Keith D. Rochfort
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Colin Davenport
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Ronan P. Murphy
- School of Health & Human Performance, Dublin City University, Glasnevin, Dublin, Ireland
- Centre for Preventive Medicine, Dublin City University, Glasnevin, Dublin, Ireland
| | - Philip M. Cummins
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
- Centre for Preventive Medicine, Dublin City University, Glasnevin, Dublin, Ireland
- * E-mail:
| |
Collapse
|
13
|
Rochfort KD, Collins LE, Murphy RP, Cummins PM. Downregulation of blood-brain barrier phenotype by proinflammatory cytokines involves NADPH oxidase-dependent ROS generation: consequences for interendothelial adherens and tight junctions. PLoS One 2014; 9:e101815. [PMID: 24992685 PMCID: PMC4081725 DOI: 10.1371/journal.pone.0101815] [Citation(s) in RCA: 194] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/11/2014] [Indexed: 12/05/2022] Open
Abstract
Background and Objectives Blood-brain barrier (BBB) dysfunction is an integral feature of neurological disorders and involves the action of multiple proinflammatory cytokines on the microvascular endothelial cells lining cerebral capillaries. There is still however, considerable ambiguity throughout the scientific literature regarding the mechanistic role(s) of cytokines in this context, thereby warranting a comprehensive in vitro investigation into how different cytokines may cause dysregulation of adherens and tight junctions leading to BBB permeabilization. Methods The present study employs human brain microvascular endothelial cells (HBMvECs) to compare/contrast the effects of TNF-α and IL-6 on BBB characteristics ranging from the expression of interendothelial junction proteins (VE-cadherin, occludin and claudin-5) to endothelial monolayer permeability. The contribution of cytokine-induced NADPH oxidase activation to altered barrier phenotype was also investigated. Results In response to treatment with either TNF-α or IL-6 (0–100 ng/ml, 0–24 hrs), our studies consistently demonstrated significant dose- and time-dependent decreases in the expression of all interendothelial junction proteins examined, in parallel with dose- and time-dependent increases in ROS generation and HBMvEC permeability. Increased expression and co-association of gp91 and p47, pivotal NADPH oxidase subunits, was also observed in response to either cytokine. Finally, cytokine-dependent effects on junctional protein expression, ROS generation and endothelial permeability could all be attenuated to a comparable extent using a range of antioxidant strategies, which included ROS depleting agents (superoxide dismutase, catalase, N-acetylcysteine, apocynin) and targeted NADPH oxidase blockade (gp91 and p47 siRNA, NSC23766). Conclusion A timely and wide-ranging investigation comparing the permeabilizing actions of TNF-α and IL-6 in HBMvECs is presented, in which we demonstrate how either cytokine can similarly downregulate the expression of interendothelial adherens and tight junction proteins leading to elevation of paracellular permeability. The cytokine-dependent activation of NADPH oxidase leading to ROS generation was also confirmed to be responsible in-part for these events.
Collapse
Affiliation(s)
- Keith D Rochfort
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Laura E Collins
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Ronan P Murphy
- School of Health and Human Performance, Dublin City University, Dublin, Ireland; Centre for Preventive Medicine, Dublin City University, Dublin, Ireland
| | - Philip M Cummins
- School of Biotechnology, Dublin City University, Dublin, Ireland; Centre for Preventive Medicine, Dublin City University, Dublin, Ireland
| |
Collapse
|