1
|
Qu H, Liu Y, Connolly JJ, Mentch FD, Kao C, Hakonarson H. Risk of Alzheimer's disease in Down syndrome: Insights gained by multi-omics. Alzheimers Dement 2025; 21:e14604. [PMID: 40207399 PMCID: PMC11982707 DOI: 10.1002/alz.14604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 04/11/2025]
Abstract
Individuals with Down syndrome (DS) are highly susceptible to Alzheimer's disease (AD). The integration of genomics, transcriptomics, epigenomics, proteomics, and metabolomics enables unprecedented understanding of DS-AD, offering a detailed picture of this complex issue. The vast -omics data also present challenges that reflect the complexity of genetic information flow. These studies nonetheless reveal critical mechanisms behind AD risk, including unique observations in DS that differ from those seen in the general population and familial dominant AD. In addition, the correlations between the AD polygenic risk score and proteins related to female infertility and autoimmune thyroiditis corroborate clinical observations. Metabolomic data reveal disrupted metabolic networks, offering prospects for a dynamic score to create specialized nutritional interventions. By adopting a multidimensional perspective with integrated reductionism, the evolving landscape presents an opportunity to identify promising directions for developing precision strategies to mitigate the impact of AD in the DS population. HIGHLIGHTS: Individuals with Down syndrome (DS) are highly susceptible to Alzheimer's disease (AD). DS-AD is characterized by its polygenic nature, extending beyond chromosome 21 with significant contributions from various chromosomes. DS-AD also presents unique features that differ from those observed in the general population and familial dominant AD. Our review consolidates key findings from genomics, transcriptomics, epigenomics, proteomics, and metabolomics, providing a comprehensive view of the molecular mechanisms underlying DS-AD. We highlight promising research directions to further elucidate the pathogenesis of DS-AD.
Collapse
Affiliation(s)
- Hui‐Qi Qu
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Yichuan Liu
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - John J. Connolly
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Frank D. Mentch
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Charlly Kao
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Hakon Hakonarson
- The Center for Applied GenomicsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Department of Pediatrics, The Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Human GeneticsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Division of Pulmonary MedicineChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Faculty of MedicineUniversity of IcelandReykjavikIceland
| |
Collapse
|
2
|
Hamad I, Sepic S, Moztarzadeh S, García-Ponce A, Waschke J, Radeva MY. Plakoglobin does not participate in endothelial barrier stabilization mediated by cAMP. Sci Rep 2025; 15:9043. [PMID: 40091082 PMCID: PMC11911453 DOI: 10.1038/s41598-025-93756-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
Critical for maintenance of endothelial barrier is the remodeling of the actin cytoskeleton and the precise control of junctional integrity. Plakoglobin (PG) is a structural and signaling protein involved in vascular permeability regulation together with key signaling molecules such as cAMP, Rho GTPases and actin-binding proteins. Here, we investigated the role of PG in cAMP-mediated endothelial barrier stabilization by establishing myocardial endothelial cells derived from wild type (WT) and PG knock-out (PG-KO) mice. Under basal conditions, TEER measurements showed increased barrier function of PG-KO, an effect associated with enhanced protein levels and junctional VE-cadherin and β-catenin accumulation. PG-KO cells also displayed more PECAM-1 and VE-PTP-phosphatase and less phosphorylated VE-cadherin, typically linked with modulation of junctional integrity. PG ablation neither changed the composition of VE-cadherin/β-catenin complex nor activities of Rac1 and RhoA but decreased the basal intracellular cAMP concentration. Remarkably, cAMP augmentation led to enhanced Rac1 activity and TEER in both cell lines, but the effect was less prominent in PG-KO. The tighter barrier in WT was paralleled with more VE-cadherin, β-catenin and cortactin, an actin-binding protein, towards junctions. Surprisingly, PG phosphorylation at Ser665 was not required for cAMP-mediated endothelial barrier integrity, which is different to cardiomyocyte and keratinocyte cell adhesion.
Collapse
Affiliation(s)
- Ibrahim Hamad
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 11, 80336, Munich, Germany
| | - Sara Sepic
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 11, 80336, Munich, Germany
| | - Sina Moztarzadeh
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 11, 80336, Munich, Germany
| | - Alexander García-Ponce
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 11, 80336, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 11, 80336, Munich, Germany
| | - Mariya Y Radeva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 11, 80336, Munich, Germany.
| |
Collapse
|
3
|
Li H, Wang R, Xu P, Yuan C, Huang M, Jiang L. Elucidating the molecular basis of PECAM-1 and Tie2 interaction from binding dynamics and complex formation. Biochem Biophys Res Commun 2024; 735:150484. [PMID: 39094232 DOI: 10.1016/j.bbrc.2024.150484] [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: 06/04/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Endothelial hyperpermeability-induced vascular dysfunction is a prevalent and significant characteristic in critical illnesses such as sepsis and other conditions marked by acute systemic inflammation. Platelet endothelial cell adhesion molecule-1 (PECAM-1) and Tie2 serve as transmembrane receptors within endothelial cells (ECs), playing pivotal roles not only in maintaining EC-EC junctions but also in influencing vasculogenesis, vessel homeostasis, and vascular remodeling. OBJECTIVES At present, the molecular basis of the PECAM-1-Tie2 interaction remains inadequately elucidated. In the study, recombinant soluble PECAM-1 (sPECAM-1) and Tie2 (sTie2) were expressed by Drosophila S2 and HEK293 expression systems, respectively. The interactions between sPECAM-1 and sTie2 were investigated using the Surface Plasmon Resonance (SPR) and size-exclusion chromatography methods. An immunofluorescence assay was used to detect the binding of sPECAM-1 and sTie2 on endothelial cells. RESULTS PECAM-1 was found to bind with sTie2 in a sodium and pH-dependent manner as confirmed by the ELISA, the D5-D6 domains of PECAM-1 might play a crucial role in binding with sTie2. Surface Plasmon Resonance (SPR) results showed that the full length of sPECAM-1 has the strongest binding affinity (KD = 48.4 nM) with sTie2, compared to sPECAM-1-D1-D4 and sPECAM-1-D1-D2. This result is consistent with that in the ELISA. In addition, size-exclusion chromatography demonstrated that sPECAM-1, sTie2, and Ang1 can form a ternary complex. CONCLUSION In this study, we determined that sPECAM-1 binds to sTie2 in a pH and sodium-dependent manner. The full length of sPECAM-1 has the strongest binding affinity, and the D5-D6 domains in sPECAM-1 play a crucial role in the interaction between sPECAM-1 and sTie2.
Collapse
Affiliation(s)
- Hao Li
- College of Chemistry, Fuzhou University, Fuzhou, 350116, China
| | - Rui Wang
- College of Chemistry, Fuzhou University, Fuzhou, 350116, China
| | - Peng Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, 350116, China; The National & Local Joint Engineering Research Center on Biopharmaceutical and Photodynamic Therapy Technologies, Fuzhou University, Fuzhou, Fujian, 350116, China.
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou, 350116, China; The National & Local Joint Engineering Research Center on Biopharmaceutical and Photodynamic Therapy Technologies, Fuzhou University, Fuzhou, Fujian, 350116, China.
| |
Collapse
|
4
|
Mariappan V, Shanmugam L, Ranganathan Green S, Easow JM, Mutheneni SR, Thirugnanasambandhar Sivasubramanian A, Balakrishna Pillai A. Increased shedding of PECAM-1 associated with elevated serum MMP-14 levels as new blood indicators of dengue disease manifestation. Infect Dis Now 2024; 54:104964. [PMID: 39181201 DOI: 10.1016/j.idnow.2024.104964] [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: 12/22/2023] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024]
Abstract
OBJECTIVES Host factors that regulate plasma leakage during severe dengue (SD) are under investigation. While PECAM-1 and MMP-14 have been reported to regulate vascular integrity, their role in dengue pathogenesis remains unexplored. This study aims to assess the association of soluble PECAM-1 and MMP-14 with dengue severity symptoms. PATIENTS AND METHODS Serum levels of PECAM-1 and MMP-14 were evaluated in dengue (N-25) comprising 10 severe dengue (SD) and 15 non-severe dengue, 10 other febrile illnesses along with healthy controls (N-10) using ELISA. Protein levels were assessed using in vitro models. RESULTS From febrile to critical phase, a significant increase in PECAM-1 (P≤0.01) & MMP-14 (P≤0.001) levels were observed in SD cases compared to non-severe or other controls. Serum levels of PECAM-1 and MMP 14 were found to be positively (P≤0.001) associated. Soluble PECAM-1 levels of severe defervescence showed a positive correlation (P≤0.001) with plasma leakage and an inverse relationship (P≤0.001) with platelet count. In vitro analysis revealed elevated expression of study proteins in endothelial cells activated with severe serum samples. To the best of our knowledge, this is the first report to explore PECAM-1 or MMP-14 dynamics and their association with dengue severity. CONCLUSION Higher shedding of sPECAM-1 accompanied with increased levels of MMP-14 is strongly associated with severe dengue. However, the exact role of serum PECAM-1 in disease prognosis requires further studies.
Collapse
Affiliation(s)
- Vignesh Mariappan
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Lokesh Shanmugam
- ICMR-National Institute of Epidemiology (ICMR-NIE), Ayapakkam, Chennai 600 070, India.
| | - Siva Ranganathan Green
- Department of General Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Joshy M Easow
- Department of Microbiology, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Srinivasa Rao Mutheneni
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, Telangana 500 007, India.
| | | | - Agieshkumar Balakrishna Pillai
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| |
Collapse
|
5
|
Memari E, Helfield B. Shear stress preconditioning and microbubble flow pattern modulate ultrasound-assisted plasma membrane permeabilization. Mater Today Bio 2024; 27:101128. [PMID: 38988819 PMCID: PMC11234154 DOI: 10.1016/j.mtbio.2024.101128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/31/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024] Open
Abstract
The recent and exciting success of anti-inflammatory therapies for ischemic heart disease (e.g. atherosclerosis) is hindered by the lack of site-specific and targeted therapeutic deposition. Microbubble-mediated focused ultrasound, which uses circulating, lipid-encapsulated intravascular microbubbles to locally enhance endothelial permeability, offers an exciting approach. Atherosclerotic plaques preferentially develop in regions with disturbed blood flow, and microbubble-endothelial cell membrane interactions under such flow conditions are not well understood. Here, using an acoustically-coupled microscopy system, endothelial cells were sonicated (1 MHz, 20 cycle bursts, 1 ms PRI, 4 s duration, 300 kPa peak-negative pressure) under perfusion with Definity™ bubbles to examine microbubble-mediated endothelial permeabilization under a range of physiological conditions. Endothelial preconditioning under prolonged shear influenced physiology and the secretome, inducing increased expression of pro-angiogenesis analytes, decreasing levels of pro-inflammatory ones, and increasing the susceptibility of ultrasound therapy. Ultrasound treatment efficiency was positively correlated with concentrations of pro-angiogenic cytokines (e.g. VEGF-A, EGF, FGF-2), and negatively correlated with pro-inflammatory chemokines (e.g. MCP-1, GCP-2, SDF-1). Furthermore, ultrasound therapy under non-reversing pulsatile flow (∼4-8 dyne/cm2, 0.5-1 Hz) increased permeabilization up to 2.4-fold compared to shear-matched laminar flow, yet treatment under reversing oscillatory flow resulted in more heterogeneous modulation. This study provides insight into the role of vascular physiology, including endothelial biology, into the design of a localized ultrasound drug delivery system for ischemic heart disease.
Collapse
Affiliation(s)
- Elahe Memari
- Department of Physics, Concordia University, Montreal, H4B 1R6, Canada
| | - Brandon Helfield
- Department of Physics, Concordia University, Montreal, H4B 1R6, Canada
- Department of Biology, Concordia University, Montreal, H4B 1R6, Canada
| |
Collapse
|
6
|
Lu Z, Ding L, Jiang X, Zhang S, Yan M, Yang G, Tian X, Wang Q. Single-nucleus RNA transcriptome profiling reveals murine adipose tissue endothelial cell proliferation gene networks involved in obesity development. Arch Biochem Biophys 2024; 757:110029. [PMID: 38729594 DOI: 10.1016/j.abb.2024.110029] [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: 01/10/2024] [Revised: 03/18/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
Endothelial cells play an important role in the metabolism of adipose tissue (AT). This study aimed to analyze the changes that adipose tissue in AT endothelial cells undergo during the development of obesity, using single-nucleus RNA sequence (snRNA-seq). Mouse paraepididymal AT cells were subjected to snRNA-seq with the 10X Genomics platform. The cell types were then clustered using t-distributed stochastic neighbor embedding and unbiased computational informatics analyses. Protein-protein interactions network was established using the STRING database and visualized using Cytoscape. The dataset was subjected to differential gene enrichment analysis. In total, 21,333 cells acquired from 24 mouse paraepididymal AT samples were analyzed using snRNA-seq. This study identified 18 distinct clusters and annotated macrophages, fibroblasts, epithelial cells, T cells, endothelial cells, stem cells, neutrophil cells, and neutrophil cell types based on representative markers. Cluster 12 was defined as endothelial cells. The proportion of endothelial cells decreased with the development of obesity. Inflammatory factors, such as Vegfa and Prdm16 were upregulated in the medium obesity group but downregulated in the obesity group. Genes, such as Prox1, Erg, Flt4, Kdr, Flt1, and Pecam1 promoted the proliferation of AT endothelial cells and maintained the internal environment of AT. This study established a reference model and general framework for studying the mechanisms, biomarkers, and therapeutic targets of endothelial cell dysfunction-related diseases at the single-cell level.
Collapse
Affiliation(s)
- Zhimin Lu
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China
| | - Ling Ding
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China
| | - Xing Jiang
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China
| | - Sen Zhang
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China
| | - Min Yan
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China
| | - Guangxin Yang
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China
| | - Xuewen Tian
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China.
| | - Qinglu Wang
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China.
| |
Collapse
|
7
|
Wang J, Lai DA, Wang JJ, Zhang SX. Effects of Nox4 upregulation on PECAM-1 expression in a mouse model of diabetic retinopathy. PLoS One 2024; 19:e0303010. [PMID: 38748682 PMCID: PMC11095704 DOI: 10.1371/journal.pone.0303010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/17/2024] [Indexed: 05/19/2024] Open
Abstract
Diabetic Retinopathy (DR) is the leading cause of vision loss in working-age adults. The hallmark features of DR include vascular leakage, capillary loss, retinal ischemia, and aberrant neovascularization. Although the pathophysiology is not fully understood, accumulating evidence supports elevated reactive oxygen species associated with increased activity of NADPH oxidase 4 (Nox4) as major drivers of disease progression. Previously, we have shown that Nox4 upregulation in retinal endothelial cells by diabetes leads to increased vascular leakage by an unknown mechanism. Platelet endothelial cell adhesion molecule 1 (PECAM-1) is a cell surface molecule that is highly expressed in endothelial cells and regulates endothelial barrier function. In the present study, using endothelial cell-specific human Nox4 transgenic (TG) mice and endothelial cell-specific Nox4 conditional knockout (cKO) mice, we investigated the impact of Nox4 upregulation on PECAM-1 expression in mouse retinas and brain microvascular endothelial cells (BMECs). Additionally, cultured human retinal endothelial cells (HRECs) transduced with adenovirus overexpressing human Nox4 were used in the study. We found that overexpression of Nox4 increases PECAM-1 mRNA but has no effect on its protein expression in the mouse retina, BMECs, or HRECs. Furthermore, PECAM-1 mRNA and protein expression was unchanged in BMECs isolated from cKO mice compared to wild type (WT) mice with or without 2 months of diabetes. Together, these findings do not support a significant role of Nox4 in the regulation of PECAM-1 expression in the diabetic retina and endothelial cells. Further studies are warranted to elucidate the mechanism of Nox4-induced vascular leakage by investigating other intercellular junctional proteins in endothelial cells and their implications in the pathophysiology of diabetic retinopathy.
Collapse
Affiliation(s)
- Jinli Wang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States of America
| | - Daniel A. Lai
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States of America
| | - Joshua J. Wang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States of America
| | - Sarah X. Zhang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States of America
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States of America
| |
Collapse
|
8
|
Siddiqui MT, Han Y, Shapiro D, West G, Fiocchi C, Cresci GAM. The Postbiotic Butyrate Mitigates Gut Mucosal Disruption Caused by Acute Ethanol Exposure. Int J Mol Sci 2024; 25:1665. [PMID: 38338944 PMCID: PMC10855591 DOI: 10.3390/ijms25031665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/12/2024] Open
Abstract
We aimed to test how the postbiotic butyrate impacts select gut bacteria, small intestinal epithelial integrity, and microvascular endothelial activation during acute ethanol exposure in mice and primary human intestinal microvascular endothelial cells (HIMECs). Supplementation during an acute ethanol challenge with or without tributyrin, a butyrate prodrug, was delivered to C57BL/6 mice. A separate group of mice received 3 days of clindamycin prior to the acute ethanol challenge. Upon euthanasia, blood endotoxin, cecal bacteria, jejunal barrier integrity, and small intestinal lamina propria dendritic cells were assessed. HIMECs were tested for activation following exposure to ethanol ± lipopolysaccharide (LPS) and sodium butyrate. Tributyrin supplementation protected a butyrate-generating microbe during ethanol and antibiotic exposure. Tributyrin rescued ethanol-induced disruption in jejunal epithelial barrier, elevated plasma endotoxin, and increased mucosal vascular addressin cell-adhesion molecule-1 (MAdCAM-1) expression in intestinal microvascular endothelium. These protective effects of tributyrin coincided with a tolerogenic dendritic response in the intestinal lamina propria. Lastly, sodium butyrate pre- and co-treatment attenuated the direct effects of ethanol and LPS on MAdCAM-1 induction in the HIMECs from a patient with ulcerative colitis. Tributyrin supplementation protects small intestinal epithelial and microvascular barrier integrity and modulates microvascular endothelial activation and dendritic tolerizing function during a state of gut dysbiosis and acute ethanol challenge.
Collapse
Affiliation(s)
- Mohamed Tausif Siddiqui
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (M.T.S.); (C.F.)
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yingchun Han
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - David Shapiro
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Gail West
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Claudio Fiocchi
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (M.T.S.); (C.F.)
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Gail A. M. Cresci
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (M.T.S.); (C.F.)
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Cleveland Clinic Children’s Hospital, Cleveland, OH 44195, USA
| |
Collapse
|
9
|
Locatelli M, Rottoli D, Mahmoud R, Abbate M, Corna D, Cerullo D, Tomasoni S, Remuzzi G, Zoja C, Benigni A, Macconi D. Endothelial Glycocalyx of Peritubular Capillaries in Experimental Diabetic Nephropathy: A Target of ACE Inhibitor-Induced Kidney Microvascular Protection. Int J Mol Sci 2023; 24:16543. [PMID: 38003732 PMCID: PMC10671403 DOI: 10.3390/ijms242216543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Peritubular capillary rarefaction is a recurrent aspect of progressive nephropathies. We previously found that peritubular capillary density was reduced in BTBR ob/ob mice with type 2 diabetic nephropathy. In this model, we searched for abnormalities in the ultrastructure of peritubular capillaries, with a specific focus on the endothelial glycocalyx, and evaluated the impact of treatment with an angiotensin-converting enzyme inhibitor (ACEi). Mice were intracardially perfused with lanthanum to visualise the glycocalyx. Transmission electron microscopy analysis revealed endothelial cell abnormalities and basement membrane thickening in the peritubular capillaries of BTBR ob/ob mice compared to wild-type mice. Remodelling and focal loss of glycocalyx was observed in lanthanum-stained diabetic kidneys, associated with a reduction in glycocalyx components, including sialic acids, as detected through specific lectins. ACEi treatment preserved the endothelial glycocalyx and attenuated the ultrastructural abnormalities of peritubular capillaries. In diabetic mice, peritubular capillary damage was associated with an enhanced tubular expression of heparanase, which degrades heparan sulfate residues of the glycocalyx. Heparanase was also detected in renal interstitial macrophages that expressed tumor necrosis factor-α. All these abnormalities were mitigated by ACEi. Our findings suggest that, in experimental diabetic nephropathy, preserving the endothelial glycocalyx is important in order to protect peritubular capillaries from damage and loss.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, 24126 Bergamo, Italy; (M.L.); (D.R.); (R.M.); (M.A.); (D.C.); (D.C.); (S.T.); (G.R.); (C.Z.); (D.M.)
| | | |
Collapse
|
10
|
Mutgan AC, Jandl K, Radic N, Valzano F, Kolb D, Hoffmann J, Foris V, Wilhelm J, Boehm PM, Hoetzenecker K, Olschewski A, Olschewski H, Heinemann A, Wygrecka M, Marsh LM, Kwapiszewska G. Pentastatin, a matrikine of the collagen IVα5, is a novel endogenous mediator of pulmonary endothelial dysfunction. Am J Physiol Cell Physiol 2023; 325:C1294-C1312. [PMID: 37694286 PMCID: PMC11550886 DOI: 10.1152/ajpcell.00391.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023]
Abstract
Deposition of basement membrane components, such as collagen IVα5, is associated with altered endothelial cell function in pulmonary hypertension. Collagen IVα5 harbors a functionally active fragment within its C-terminal noncollageneous (NC1) domain, called pentastatin, whose role in pulmonary endothelial cell behavior remains unknown. Here, we demonstrate that pentastatin serves as a mediator of pulmonary endothelial cell dysfunction, contributing to pulmonary hypertension. In vitro, treatment with pentastatin induced transcription of immediate early genes and proinflammatory cytokines and led to a functional loss of endothelial barrier integrity in pulmonary arterial endothelial cells. Mechanistically, pentastatin leads to β1-integrin subunit clustering and Rho/ROCK activation. Blockage of the β1-integrin subunit or the Rho/ROCK pathway partially attenuated the pentastatin-induced endothelial barrier disruption. Although pentastatin reduced the viability of endothelial cells, smooth muscle cell proliferation was induced. These effects on the pulmonary vascular cells were recapitulated ex vivo in the isolated-perfused lung model, where treatment with pentastatin-induced swelling of the endothelium accompanied by occasional endothelial cell apoptosis. This was reflected by increased vascular permeability and elevated pulmonary arterial pressure induced by pentastatin. This study identifies pentastatin as a mediator of endothelial cell dysfunction, which thus might contribute to the pathogenesis of pulmonary vascular disorders such as pulmonary hypertension.NEW & NOTEWORTHY This study is the first to show that pentastatin, the matrikine of the basement membrane (BM) collagen IVα5 polypeptide, triggers rapid pulmonary arterial endothelial cell barrier disruption, activation, and apoptosis in vitro and ex vivo. Mechanistically, pentastatin partially acts through binding to the β1-integrin subunit and the Rho/ROCK pathway. These findings are the first to link pentastatin to pulmonary endothelial dysfunction and, thus, suggest a major role for BM-matrikines in pulmonary vascular diseases such as pulmonary hypertension.
Collapse
Affiliation(s)
- Ayse Ceren Mutgan
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Katharina Jandl
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Nemanja Radic
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Francesco Valzano
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Dagmar Kolb
- Core Facility Ultrastructure Analysis, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Julia Hoffmann
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Vasile Foris
- Division of Pulmonology, Medical University of Graz, Graz, Austria
| | - Jochen Wilhelm
- Institute for Lung Health, Member of the German Lung Center (DZL), Giessen, Germany
| | - Panja M Boehm
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Division of Pulmonology, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Malgorzata Wygrecka
- Institute for Lung Health, Member of the German Lung Center (DZL), Giessen, Germany
- Center for Infection and Genomics of the Lung, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Leigh M Marsh
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Grazyna Kwapiszewska
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Institute for Lung Health, Member of the German Lung Center (DZL), Giessen, Germany
| |
Collapse
|
11
|
Chen SN, Mai ZY, Mai JN, Liang W, Dong ZX, Ju FE, Chan SH, Fang Z, Xu Y, Uziel O, He C, Zhang XD, Zheng Y. E2F2 modulates cell adhesion through the transcriptional regulation of PECAM1 in multiple myeloma. Br J Haematol 2023; 202:840-855. [PMID: 37365680 DOI: 10.1111/bjh.18958] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/26/2023] [Accepted: 06/17/2023] [Indexed: 06/28/2023]
Abstract
Multiple myeloma (MM) is the second most common haematological malignancy. Despite the development of new drugs and treatments in recent years, the therapeutic outcomes of patients are not satisfactory. It is necessary to further investigate the molecular mechanism underlying MM progression. Herein, we found that high E2F2 expression was correlated with poor overall survival and advanced clinical stages in MM patients. Gain- and loss-of-function studies showed that E2F2 inhibited cell adhesion and consequently activated cell epithelial-to-mesenchymal transition (EMT) and migration. Further experiments revealed that E2F2 interacted with the PECAM1 promoter to suppress its transcriptional activity. The E2F2-knockdown-mediated promotion of cell adhesion was significantly reversed by the repression of PECAM1 expression. Finally, we observed that silencing E2F2 significantly inhibited viability and tumour progression in MM cell models and xenograft mouse models respectively. This study demonstrates that E2F2 plays a vital role as a tumour accelerator by inhibiting PECAM1-dependent cell adhesion and accelerating MM cell proliferation. Therefore, E2F2 may serve as a potential independent prognostic marker and therapeutic target for MM.
Collapse
Affiliation(s)
- Shu-Na Chen
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Zhi-Ying Mai
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Jun-Na Mai
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Weiyao Liang
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Zhao-Xia Dong
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Fei-Er Ju
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Sze-Hoi Chan
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Zhigang Fang
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yichuan Xu
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Orit Uziel
- The Felsenstein Medical Research Center, Institute of Hematology Rabin Medical Center and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Xing-Ding Zhang
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Yongjiang Zheng
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
12
|
Adzraku SY, Wang G, Cao C, Bao Y, Wang Y, Smith AO, Du Y, Wang H, Li Y, Xu K, Qiao J, Ju W, Zeng L. Robo4 inhibits gamma radiation-induced permeability of a murine microvascular endothelial cell by regulating the junctions. Cell Mol Biol Lett 2023; 28:2. [PMID: 36647012 PMCID: PMC9843922 DOI: 10.1186/s11658-022-00413-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/19/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Hematopoietic stem cell transplantation involves irradiation preconditioning which causes bone marrow endothelial cell dysfunction. While much emphasis is on the reconstitution of hematopoietic stem cells in the bone marrow microenvironment, endothelial cell preservation is indispensable to overcome the preconditioning damages. This study aims to ascertain the role of Roundabout 4 (Robo4) in regulating irradiation-induced damage to the endothelium. METHODS Microvascular endothelial cells were treated with γ-radiation to establish an endothelial cell injury model. Robo4 expression in the endothelial cells was manipulated employing lentiviral-mediated RNAi and gene overexpression technology before irradiation treatment. The permeability of endothelial cells was measured using qPCR, immunocytochemistry, and immunoblotting to analyze the effect on the expression and distribution of junctional molecules, adherens junctions, tight junctions, and gap junctions. Using Transwell endothelial monolayer staining, FITC-Dextran permeability, and gap junction-mediated intercellular communication (GJIC) assays, we determined the changes in endothelial functions after Robo4 gene manipulation and irradiation. Moreover, we measured the proportion of CD31 expression in endothelial cells by flow cytometry. We analyzed variations between two or multiple groups using Student's t-tests and ANOVA. RESULTS Ionizing radiation upregulates Robo4 expression but disrupts endothelial junctional molecules. Robo4 deletion causes further degradation of endothelial junctions hence increasing the permeability of the endothelial cell monolayer. Robo4 knockdown in microvascular endothelial cells increases the degradation and delocalization of ZO-1, PECAM-1, occludin, and claudin-5 molecules after irradiation. Conversely, connexin 43 expression increases after silencing Robo4 in endothelial cells to induce permeability but are readily destroyed when exposed to 10 Gy of gamma radiation. Also, Robo4 knockdown enhances Y731-VE-cadherin phosphorylation leading to the depletion and destabilization of VE-cadherin at the endothelial junctions following irradiation. However, Robo4 overexpression mitigates irradiation-induced degradation of tight junctional proteins and stabilizes claudin-5 and ZO-1 distribution. Finally, the enhanced expression of Robo4 ameliorates the irradiation-induced depletion of VE-cadherin and connexin 43, improves the integrity of microvascular endothelial cell junctions, and decreases permeability. CONCLUSION This study reveals that Robo4 maintains microvascular integrity after radiation preconditioning treatment by regulating endothelial permeability and protecting endothelial functions. Our results also provided a potential mechanism to repair the bone marrow vascular niche after irradiation by modulating Robo4 expression.
Collapse
Affiliation(s)
- Seyram Yao Adzraku
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China ,Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002 China
| | - Guozhang Wang
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China ,Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002 China
| | - Can Cao
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China ,Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002 China
| | - Yurong Bao
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Yizhou Wang
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Alhaji Osman Smith
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Yuwei Du
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Haiyang Wang
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Yue Li
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Kailin Xu
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Jianlin Qiao
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Wen Ju
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China ,Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002 China
| | - Lingyu Zeng
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| |
Collapse
|
13
|
Wang Y, Wang C, Zuo N, Yang H, Fang S, Shi J. Extracellular Traps Increase Burden of Bleeding by Damaging Endothelial Cell in Acute Promyelocytic Leukaemia. Front Immunol 2022; 13:841445. [PMID: 35479063 PMCID: PMC9035902 DOI: 10.3389/fimmu.2022.841445] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
The rate of complete remission of acute promyelocytic leukemia (APL) is currently over 90% because of the use of all-trans retinoic acid (ATRA) with arsenic trioxide (ATO). However, hemorrhagic mortality has emerged as the most significant barrier to APL-induced remission. Neutrophils extracellular traps (NETs/ETs) cause vascular leakage by damaging the integrity of endothelial cells. We have previously demonstrated that APL cells treated with ATRA/ATO undergo a cell death process, releasing extracellular chromatin, termed ETosis/NETosis. However, the mechanism underlying the involvement of ETs in endothelial injury in APL remain largely unknown. Here, we analysed the ability of mature and immature neutrophils to release ETs, and their interaction with platelets (PLTs) in APL. Importantly, the effect of ETs on vascular endothelium in APL was discussed. Our results showed that the ability of immature neutrophils to release ETs was impaired in APL, whereas mature neutrophils produced ETs, which were associated with activated PLTs. Moreover, ATRA+ATO induced immature neutrophil differentiation, as well as increased the release of ETs from mature neutrophils. The excessive ETs damaged endothelial cells, causing blood cell leakage. Removing ETs using DNase 1 alleviated endothelial damage and improved blood cells leakage. Our results indicate that vascular endothelial injury is at least partially associated with ETs in APL, and that targeting ETs production may be an effective approach for relieving vascular leakage and reducing the burden of bleeding in APL.
Collapse
Affiliation(s)
- Yufeng Wang
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Chunxu Wang
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Nan Zuo
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao Yang
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
- Department of General Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shaohong Fang
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Jialan Shi, , ; Shaohong Fang,
| | - Jialan Shi
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin, China
- Departments of Research and Medical Oncology, Veterans Affairs (VA) Boston Healthcare System, Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA, United States
- *Correspondence: Jialan Shi, , ; Shaohong Fang,
| |
Collapse
|
14
|
Liao D, Sundlov J, Zhu J, Mei H, Hu Y, Newman DK, Newman PJ. Atomic Level Dissection of the Platelet Endothelial Cell Adhesion Molecule 1 (PECAM-1) Homophilic Binding Interface: Implications for Endothelial Cell Barrier Function. Arterioscler Thromb Vasc Biol 2022; 42:193-204. [PMID: 34937389 PMCID: PMC8942131 DOI: 10.1161/atvbaha.121.316668] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE PECAM-1 (platelet endothelial cell adhesion molecule 1) is a 130 kDa member of the immunoglobulin (Ig) gene superfamily that is expressed on the surfaces of platelets and leukocytes and concentrated at the intercellular junctions of confluent endothelial cell monolayers. PECAM-1 Ig domains 1 and 2 (IgD1 and IgD2) engage in homophilic interactions that support a host of vascular functions, including support of leukocyte transendothelial migration and the maintenance of endothelial junctional integrity. The recently solved crystal structure of PECAM-1 IgD1 and IgD2 revealed a number of intermolecular interfaces predicted to play important roles in stabilizing PECAM-1/PECAM-1 homophilic interactions and in formation and maintenance of endothelial cell-cell contacts. We sought to determine whether the protein interfaces implicated in the crystal structure reflect physiologically important interactions. Approach and Results: We assessed the impact of single amino acid substitutions at the interfaces between opposing PECAM-1 molecules on homophilic binding and endothelial cell function. Substitution of key residues within the IgD1-IgD1 and IgD1-IgD2 interfaces but not those within the smaller IgD2-IgD2 interface, markedly disrupted PECAM-1 homophilic binding and its downstream effector functions, including the ability of PECAM-1 to localize at endothelial cell-cell borders, mediate the formation of endothelial tubes, and restore endothelial barrier integrity. CONCLUSIONS Taken together, these results validate the recently described PECAM-1 IgD1/IgD2 crystal structure by demonstrating that specific residues visualized within the IgD1-IgD1 and IgD1-IgD2 interfaces of opposing molecules in the crystal are required for functionally important homophilic interactions. This information can now be exploited to modulate functions of PECAM-1 in vivo.
Collapse
Affiliation(s)
- Danying Liao
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI,Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jesse Sundlov
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI
| | - Jieqing Zhu
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI,Department of Biochemistry, Medical College of Wisconsin, Milwaukee
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Debra K. Newman
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI,Department of Pharmacology, Medical College of Wisconsin, Milwaukee,Department of Microbiology Medical College of Wisconsin, Milwaukee,Department of The Cardiovascular Center, Medical College of Wisconsin, Milwaukee
| | - Peter J. Newman
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI,Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China,Department of Pharmacology, Medical College of Wisconsin, Milwaukee,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee,Department of The Cardiovascular Center, Medical College of Wisconsin, Milwaukee
| |
Collapse
|
15
|
Kosiński K, Malinowski D, Safranow K, Dziedziejko V, Pawlik A. PECAM1, COL4A2, PHACTR1, and LMOD1 Gene Polymorphisms in Patients with Unstable Angina. J Clin Med 2022; 11:jcm11020373. [PMID: 35054067 PMCID: PMC8778316 DOI: 10.3390/jcm11020373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 11/16/2022] Open
Abstract
Coronary artery disease (CAD) is a syndrome resulting from myocardial ischaemia of heterogeneous pathomechanism. Environmental and genetic factors contribute to its development. Atherosclerotic plaques that significantly narrow the lumen of coronary arteries cause symptoms of myocardial ischaemia. Acute coronary incidents are most often associated with plaque rupture or erosion accompanied by local activation of the coagulation system with thrombus formation. Plaque formation and stability are influenced by endothelial function and vascular smooth muscle cell function. In this study, we investigated the association between polymorphisms in genes affecting endothelial and vascular smooth muscle cell (VSMC) function and the occurrence of unstable angina pectoris. The aim of this study was to evaluate the association between the PECAM1 (rs1867624), COL4A2 (rs4773144), PHACTR1 (rs9349379) and LMOD1 (rs2820315) gene polymorphisms and the risk of unstable angina. The study included 232 patients with unstable angina diagnosed on the basis of clinical symptoms and coronary angiography and 144 healthy subjects with no significant coronary lumen stenosis at coronary angiography. There were no statistically significant differences in the distribution of COL4A2 rs4773144 and PECAM1 rs1867624 gene polymorphisms between patients with unstable angina and control subjects. In patients with unstable angina, there was an increased frequency of PHACTR1 rs9349379 G allele carriers (GG and AG genotypes) (GG+AG vs. AA, OR 1.71; 95% CI 1.10-2.66, p = 0.017) and carriers of the LMOD1 rs2820315 T allele (TT and CT genotypes) (TT+CT vs. CC, OR 1.65; 95% CI 1.09-2.51, p = 0.019) compared to the control group. The association between these alleles and unstable angina was confirmed by multivariate logistic regression analysis, in which the number of G (PHACTR1 rs9349379) and T (LMOD1 rs2820315) alleles was an independent risk factor for unstable angina. The results suggest an association between PHACTR1 rs9349379 and LMOD1 rs2820315 polymorphisms and the risk of unstable angina.
Collapse
Affiliation(s)
- Krzysztof Kosiński
- Department of Cardiology, Hospital in Szczecin, Arkonska 4, 71-455 Szczecin, Poland;
| | - Damian Malinowski
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.S.); (V.D.)
| | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.S.); (V.D.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
- Correspondence:
| |
Collapse
|
16
|
Cabrera JTO, Makino A. Efferocytosis of vascular cells in cardiovascular disease. Pharmacol Ther 2022; 229:107919. [PMID: 34171333 PMCID: PMC8695637 DOI: 10.1016/j.pharmthera.2021.107919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/21/2021] [Accepted: 06/03/2021] [Indexed: 12/20/2022]
Abstract
Cell death and the clearance of apoptotic cells are tightly regulated by various signaling molecules in order to maintain physiological tissue function and homeostasis. The phagocytic removal of apoptotic cells is known as the process of efferocytosis, and abnormal efferocytosis is linked to various health complications and diseases, such as cardiovascular disease, inflammatory diseases, and autoimmune diseases. During efferocytosis, phagocytic cells and/or apoptotic cells release signals, such as "find me" and "eat me" signals, to stimulate the phagocytic engulfment of apoptotic cells. Primary phagocytic cells are macrophages and dendritic cells; however, more recently, other neighboring cell types have also been shown to exhibit phagocytic character, including endothelial cells and fibroblasts, although they are comparatively slower in clearing dead cells. In this review, we focus on macrophage efferocytosis of vascular cells, such as endothelial cells, smooth muscle cells, fibroblasts, and pericytes, and its relation to the progression and development of cardiovascular disease. We also highlight the role of efferocytosis-related molecules and their contribution to the maintenance of vascular homeostasis.
Collapse
Affiliation(s)
- Jody Tori O Cabrera
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
17
|
Rosa A, Giese W, Meier K, Alt S, Klaus-Bergmann A, Edgar LT, Bartels E, Collins R, Szymborska A, Coxam B, Bernabeu MO, Gerhardt H. Wasp controls oriented migration of endothelial cells to achieve functional vascular patterning. Development 2021; 149:273808. [PMID: 34931661 PMCID: PMC8918813 DOI: 10.1242/dev.200195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/10/2021] [Indexed: 11/21/2022]
Abstract
Endothelial cell migration and proliferation are essential for the establishment of a hierarchical organization of blood vessels and optimal distribution of blood. However, how these cellular processes are quantitatively coordinated to drive vascular network morphogenesis remains unknown. Here, using the zebrafish vasculature as a model system, we demonstrate that the balanced distribution of endothelial cells, as well as the resulting regularity of vessel calibre, is a result of cell migration from veins towards arteries and cell proliferation in veins. We identify the Wiskott-Aldrich Syndrome protein (WASp) as an important molecular regulator of this process and show that loss of coordinated migration from veins to arteries upon wasb depletion results in aberrant vessel morphology and the formation of persistent arteriovenous shunts. We demonstrate that WASp achieves its function through the coordination of junctional actin assembly and PECAM1 recruitment and provide evidence that this is conserved in humans. Overall, we demonstrate that functional vascular patterning in the zebrafish trunk is established through differential cell migration regulated by junctional actin, and that interruption of differential migration may represent a pathomechanism in vascular malformations. Summary: Regular diameter of developing veins and arteries in the zebrafish trunk is controlled by differential endothelial cell proliferation and WASp-driven directed cell migration.
Collapse
Affiliation(s)
- André Rosa
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Wolfgang Giese
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Katja Meier
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Silvanus Alt
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Alexandra Klaus-Bergmann
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Lowell T Edgar
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Eireen Bartels
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Russell Collins
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Anna Szymborska
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Baptiste Coxam
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Miguel O Bernabeu
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK.,The Bayes Centre, The University of Edinburgh, Edinburgh, United Kingdom. 5 Berlin Institute of Health (BIH), Berlin, Germany
| | - Holger Gerhardt
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| |
Collapse
|
18
|
Yu L, Liu S, Wang C, Zhang C, Wen Y, Zhang K, Chen S, Huang H, Liu Y, Wu L, Han Z, Chen X, Li Z, Liu N. Embryonic stem cell-derived extracellular vesicles promote the recovery of kidney injury. Stem Cell Res Ther 2021; 12:379. [PMID: 34215331 PMCID: PMC8254253 DOI: 10.1186/s13287-021-02460-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/10/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Embryonic stem cell-derived extracellular vesicles (ESC-EVs) possess therapeutic potential for a variety of diseases and are considered as an alternative of ES cells. Acute kidney injury (AKI) is a common acute and severe disease in clinical practice, which seriously threatens human life and health. However, the roles and mechanisms of ESC-EVs on AKI remain unclear. METHODS In this study, we evaluated the effects of ESC-EVs on physiological repair and pathological repair using murine ischemia-reperfusion injury-induced AKI model, the potential mechanisms of which were next investigated. EVs were isolated from ESCs and EVs derived from mouse fibroblasts as therapeutic controls. We then investigated whether ESC-EVs can restore the structure and function of the damaged kidney by promoting physiological repair and inhibiting the pathological repair process after AKI in vivo and in vitro. RESULTS We found that ESC-EVs significantly promoted the recovery of the structure and function of the damaged kidney. ESC-EVs increased the proliferation of renal tubular epithelial cells, facilitated renal angiogenesis, inhibited the progression of renal fibrosis, and rescued DNA damage caused by ischemia and reperfusion after AKI. Finally, we found that ESC-EVs play a therapeutic effect by activating Sox9+ cells. CONCLUSIONS ESC-EVs significantly promote the physiological repair and inhibit the pathological repair after AKI, enabling restoration of the structure and function of the damaged kidney. This strategy might emerge as a novel therapeutic strategy for ESC clinical application.
Collapse
Affiliation(s)
- Lu Yu
- School of Medicine, Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Siying Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Chen Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Chuanyu Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yajie Wen
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Kaiyue Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Shang Chen
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Haoyan Huang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yue Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Lingling Wu
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Zhongchao Han
- Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health & Biotech Co., Beijing, China
| | - Xiangmei Chen
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China.
| | - Zongjin Li
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Na Liu
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
19
|
Eshaq RS, Harris NR. The role of tumor necrosis factor-α and interferon-γ in the hyperglycemia-induced ubiquitination and loss of platelet endothelial cell adhesion molecule-1 in rat retinal endothelial cells. Microcirculation 2021; 28:e12717. [PMID: 34008903 PMCID: PMC10078990 DOI: 10.1111/micc.12717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 05/03/2021] [Accepted: 05/11/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVE This study aimed to investigate the role of the hyperglycemia-induced increase in tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) in the ubiquitination and degradation of platelet endothelial cell adhesion molecule-1 (PECAM-1) in the diabetic retina. METHODS Type I diabetes was induced in rats by the injection of streptozotocin, with age-matched non-diabetic rats as controls. Primary rat retinal microvascular endothelial cells were grown in normal or high glucose media for 6 days or in normal glucose media for 24 h with addition of TNF-α and/or IFN-γ. PECAM-1, TNF-α, IFN-γ, and ubiquitin levels were assessed using Western blotting, immunofluorescence, and immunoprecipitation assays. Additionally, proteasome activity was assessed both in vivo and in vitro. RESULTS Under hyperglycemic conditions, total ubiquitination levels in the retina and RRMECs, and PECAM-1 ubiquitination levels in RRMECs, were significantly increased. Additionally, TNF-α and IFN-γ levels were significantly increased under hyperglycemic conditions. PECAM-1 levels in RRMECs treated with TNF-α and/or IFN-γ were significantly decreased. Moreover, there was a significant decrease in proteasome activity in the diabetic retina, hyperglycemic RRMECs, and RRMECs treated with TNF-α or IFN-γ. CONCLUSION Tumor necrosis factor-α and IFN-γ may contribute to the hyperglycemia-induced loss of PECAM-1 in retinal endothelial cells, possibly by upregulating PECAM-1 ubiquitination.
Collapse
Affiliation(s)
- Randa S Eshaq
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Norman R Harris
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| |
Collapse
|
20
|
Wan Y, Yang S, Peng M, Gama M, Yang Z, Deng X, Zhou J, Ouyang C, Luo H. Controllable synthesis of biomimetic nano/submicro-fibrous tubes for potential small-diameter vascular grafts. J Mater Chem B 2021; 8:5694-5706. [PMID: 32510089 DOI: 10.1039/d0tb01002b] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mimicking the morphological structure of native blood vessels is critical for the development of vascular grafts. Herein, small-diameter composite vascular grafts that integrate the nanofibrous bacterial cellulose (BC) and submicrofibrous cellulose acetate (CA) were fabricated via a combined electrospinning and step-by-step in situ biosynthesis. Scanning electron microscopy (SEM) observation shows the nano/submicro-fibrous morphology and well-interconnected porous structure of the BC/CA grafts. It is found that the BC/CA graft with a suitable BC content demonstrates lower potential of thrombus formation and enhanced endothelialization as compared to the BC and CA counterparts. Western blotting and RT-qPCR results suggest that the BC/CA-2 graft promotes endothelialization by improving expressions of genes vWF-1 and CD31 and protein CD31. The in vivo tests demonstrate much lower inflammatory response to the BC/CA graft. These results suggest that the BC/CA graft shows a great potential as an artificial graft for rapid formation of an endothelial cell monolayer.
Collapse
Affiliation(s)
- Yizao Wan
- Jiangxi Key Laboratory of Nanobiomaterials, Institute of Advanced Materials, East China Jiaotong University, Nanchang 330013, China. and School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Shanshan Yang
- Jiangxi Key Laboratory of Nanobiomaterials, Institute of Advanced Materials, East China Jiaotong University, Nanchang 330013, China.
| | - Mengxia Peng
- Jiangxi Key Laboratory of Nanobiomaterials, Institute of Advanced Materials, East China Jiaotong University, Nanchang 330013, China.
| | - Miguel Gama
- Centro de Engenharia Biológica, Universidade do Minho, Campus de Gualtar, P 4715-057 Braga, Portugal
| | - Zhiwei Yang
- Jiangxi Key Laboratory of Nanobiomaterials, Institute of Advanced Materials, East China Jiaotong University, Nanchang 330013, China.
| | - Xiaoyan Deng
- Jiangxi Key Laboratory of Nanobiomaterials, Institute of Advanced Materials, East China Jiaotong University, Nanchang 330013, China. and Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Jianye Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Chenxi Ouyang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Honglin Luo
- Jiangxi Key Laboratory of Nanobiomaterials, Institute of Advanced Materials, East China Jiaotong University, Nanchang 330013, China. and School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
| |
Collapse
|
21
|
Sluiter TJ, van Buul JD, Huveneers S, Quax PHA, de Vries MR. Endothelial Barrier Function and Leukocyte Transmigration in Atherosclerosis. Biomedicines 2021; 9:328. [PMID: 33804952 PMCID: PMC8063931 DOI: 10.3390/biomedicines9040328] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/24/2022] Open
Abstract
The vascular endothelium is a highly specialized barrier that controls passage of fluids and migration of cells from the lumen into the vessel wall. Endothelial cells assist leukocytes to extravasate and despite the variety in the specific mechanisms utilized by different leukocytes to cross different vascular beds, there is a general principle of capture, rolling, slow rolling, arrest, crawling, and ultimately diapedesis via a paracellular or transcellular route. In atherosclerosis, the barrier function of the endothelium is impaired leading to uncontrolled leukocyte extravasation and vascular leakage. This is also observed in the neovessels that grow into the atherosclerotic plaque leading to intraplaque hemorrhage and plaque destabilization. This review focuses on the vascular endothelial barrier function and the interaction between endothelial cells and leukocytes during transmigration. We will discuss the role of endothelial dysfunction, transendothelial migration of leukocytes and plaque angiogenesis in atherosclerosis.
Collapse
Affiliation(s)
- Thijs J. Sluiter
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jaap D. van Buul
- Sanquin Research and Landsteiner Laboratory, Leeuwenhoek Centre for Advanced Microscopy, Swammerdam Institute for Life Sciences, University of Amsterdam, 1066 CX Amsterdam, The Netherlands;
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Paul H. A. Quax
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Margreet R. de Vries
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
22
|
Soriano Jerez EM, Gibbins JM, Hughes CE. Targeting platelet inhibition receptors for novel therapies: PECAM-1 and G6b-B. Platelets 2021; 32:761-769. [PMID: 33646086 DOI: 10.1080/09537104.2021.1882668] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
While current oral antiplatelet therapies benefit many patients, they deregulate the hemostatic balance leaving patients at risk of systemic side-effects such as hemorrhage. Dual antiplatelet treatment is the standard approach, combining aspirin with P2Y12 blockers. These therapies mainly target autocrine activation mechanisms (TxA2, ADP) and, more recently, the use of thrombin or thrombin receptor antagonists have been added to the available approaches. Recent efforts to develop new classes of anti-platelet drugs have begun to focus on primary platelet activation pathways such as through the immunoreceptor tyrosine-based activation motif (ITAM)-containing collagen receptor GPVI/FcRγ-chain complex. There are already encouraging results from targeting GPVI, with reduced aggregation and smaller arterial thrombi, without major bleeding complications, likely due to overlapping activation signaling pathways with other receptors such as the GPIb-V-IX complex. An alternative approach to reduce platelet activation could be to inhibit this signaling pathway by targeting the inhibitory pathways intrinsic to platelets. Stimulation of endogenous negative modulators could provide more specific inhibition of platelet function, but is this feasible? In this review, we explore the potential of the two major platelet immunoreceptor tyrosine-based inhibitory motif (ITIM)-containing inhibitory receptors, G6b-B and PECAM-1, as antithrombotic targets.
Collapse
Affiliation(s)
- Eva M Soriano Jerez
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK.,Institute of Experimental Biomedicine, University Hospital Würzburg and Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Craig E Hughes
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| |
Collapse
|
23
|
Inflammation-Related Risk Loci in Genome-Wide Association Studies of Coronary Artery Disease. Cells 2021; 10:cells10020440. [PMID: 33669721 PMCID: PMC7921935 DOI: 10.3390/cells10020440] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/02/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
Although the importance of inflammation in atherosclerosis is now well established, the exact molecular processes linking inflammation to the development and course of the disease are not sufficiently understood. In this context, modern genetics—as applied by genome-wide association studies (GWAS)—can serve as a comprehensive and unbiased tool for the screening of potentially involved pathways. Indeed, a considerable proportion of loci discovered by GWAS is assumed to affect inflammatory processes. Despite many well-replicated association findings, however, translating genomic hits to specific molecular mechanisms remains challenging. This review provides an overview of the currently most relevant inflammation-related GWAS findings in coronary artery disease and explores their potential clinical perspectives.
Collapse
|
24
|
Rahman I, Collado Sánchez A, Davies J, Rzeniewicz K, Abukscem S, Joachim J, Hoskins Green HL, Killock D, Sanz MJ, Charras G, Parsons M, Ivetic A. L-selectin regulates human neutrophil transendothelial migration. J Cell Sci 2021; 134:jcs.250340. [PMID: 33408247 PMCID: PMC7888707 DOI: 10.1242/jcs.250340] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 12/03/2020] [Indexed: 01/13/2023] Open
Abstract
The migration of circulating neutrophils towards damaged or infected tissue is absolutely critical to the inflammatory response. L-selectin is a cell adhesion molecule abundantly expressed on circulating neutrophils. For over two decades, neutrophil L-selectin has been assigned the exclusive role of supporting tethering and rolling – the initial stages of the multi-step adhesion cascade. Here, we provide direct evidence for L-selectin contributing to neutrophil transendothelial migration (TEM). We show that L-selectin co-clusters with PECAM-1 – a well-characterised cell adhesion molecule involved in regulating neutrophil TEM. This co-clustering behaviour occurs specifically during TEM, which serves to augment ectodomain shedding of L-selectin and expedite the time taken for TEM (TTT) to complete. Blocking PECAM-1 signalling (through mutation of its cytoplasmic tail), PECAM-1-dependent adhesion or L-selectin shedding, leads to a significant delay in the TTT. Finally, we show that co-clustering of L-selectin with PECAM-1 occurs specifically across TNF- but not IL-1β-activated endothelial monolayers – implying unique adhesion interactomes forming in a cytokine-specific manner. To our knowledge, this is the first report to implicate a non-canonical role for L-selectin in regulating neutrophil TEM. Highlighted Article: Neutrophil L-selectin co-clusters with PECAM-1 in cis during transendothelial migration (TEM). Clustering neutrophil PECAM-1 activates p38 MAPK and JNK to regulate L-selectin shedding, which in turn expedites TEM.
Collapse
Affiliation(s)
- Izajur Rahman
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Aida Collado Sánchez
- Department of Pharmacology and Faculty of Medicine and Odontology, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain.,Institute of Health Research INCLIVA, University Clinic Hospital of Valencia, Av. Menéndez Pelayo 4, 46010, Valencia, Spain
| | - Jessica Davies
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Karolina Rzeniewicz
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Sarah Abukscem
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Justin Joachim
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Hannah L Hoskins Green
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - David Killock
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Maria Jesus Sanz
- Department of Pharmacology and Faculty of Medicine and Odontology, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain.,Institute of Health Research INCLIVA, University Clinic Hospital of Valencia, Av. Menéndez Pelayo 4, 46010, Valencia, Spain.,CIBERDEM-Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, ISCIII, Av. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Guillaume Charras
- London Centre for Nanotechnology, University College London, London WC1H 0AH, UK
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Aleksandar Ivetic
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
25
|
Krautter F, Iqbal AJ. Glycans and Glycan-Binding Proteins as Regulators and Potential Targets in Leukocyte Recruitment. Front Cell Dev Biol 2021; 9:624082. [PMID: 33614653 PMCID: PMC7890243 DOI: 10.3389/fcell.2021.624082] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/12/2021] [Indexed: 12/19/2022] Open
Abstract
Leukocyte recruitment is a highly controlled cascade of interactions between proteins expressed by the endothelium and circulating leukocytes. The involvement of glycans and glycan-binding proteins in the leukocyte recruitment cascade has been well-characterised. However, our understanding of these interactions and their regulation has expanded substantially in recent years to include novel lectins and regulatory pathways. In this review, we discuss the role of glycans and glycan-binding proteins, mediating the interactions between endothelium and leukocytes both directly and indirectly. We also highlight recent findings of key enzymes involved in glycosylation which affect leukocyte recruitment. Finally, we investigate the potential of glycans and glycan binding proteins as therapeutic targets to modulate leukocyte recruitment and transmigration in inflammation.
Collapse
Affiliation(s)
- Franziska Krautter
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Asif J Iqbal
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
26
|
Arciniegas E, Carrillo LM, Rojas H, Pineda J, Ramírez R, Reyes O, Chopite M, Rocheta A. Plump endothelial cells integrated into pre-existing venules contribute to the formation of 'mother' and 'daughter' vessels in pyogenic granuloma: possible role of galectin-1, -3 and -8. Scars Burn Heal 2021; 7:2059513120986687. [PMID: 33796337 PMCID: PMC7841855 DOI: 10.1177/2059513120986687] [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] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Pyogenic granuloma (PG) is a reactive inflammatory vascular lesion of the skin and mucous membranes, characterised by the presence of enlarged venules and seamed and seamless capillaries with plump endothelial cells (EC), and numerous macrophages. EC activation upregulates the synthesis of galectins and induces their translocation to the EC surface promoting angiogenesis and lymphangiogenesis, particularly galectin-1 (Gal-1), Gal-3 and Gal-8. However, the presence and distribution of Gal-1, -3 and -8, as well as their implications in the pathogenesis of PG, has not been considered. MATERIALS AND METHODS Eight biopsies from patients diagnosed with PG were selected. The presence of PECAM-1/CD31, IL-1β, VEGF-C, VEGFR-2, VEGFR-3, integrin β1, CD44, fibronectin and Gal-1, -3 and -8 was assessed by immunofluorescence staining using confocal laser scanning microscopy. RESULTS AND DISCUSSION Immunostaining revealed that these molecules were present in the enlarged venules with plump ECs, in some macrophages and other immune cells. We propose that macrophages release VEGF-A and VEGF-C inducing VEGFR-2/VEGFR-3 expression and activation, leading macrophages to transdifferentiate into plump ECs that might integrate into pre-existing venules, contributing to the formation of enlarged venules with transluminal bridges and capillaries. EC activation, induced by certain cytokines, has been shown to stimulate galectin expression and changes in the cellular localisation through association and activation of specific EC surface glycoproteins. Therefore, it is plausible that Gal-1, -3 and -8, acting in a concerted manner, could be mediating the transdifferentiation of macrophages into plump ECs and facilitating their migration and incorporation into the new vessels. LAY SUMMARY In this study, immunostaining of pyogenic granuloma (PG) tissue sections showed immunoreactivity for PECAM-1/CD31, IL-1β, VEGF-C, VEGFR-2 and VEGFR-3, and galectin-1, -3 and -8 in enlarged venules with plump endothelial cells (EC), as well as in some macrophages and other immune cells. Interestingly, enlarged and thin-walled transient vessels lined by PECAM-1/CD31 and VEGFR-2 immunopositive ECs that form from pre-existing normal venules in response to VEGF-A (called 'mother' vessels [MV]) and that undergo intraluminal bridging evolving into various types of capillaries (called 'daughter' vessels [DV]) have been observed in benign and malignant tumours, in physiological and pathological angiogenesis as well as in vascular malformations, suggesting an important role for VEGF-A and VEGFR-2 in such a process. However, it is not only the mechanisms by which the MVs evolve in different types of DVs that remains to be elucidated, but also whether the cells that form intraluminal bridges proceed from locally activated ECs or whether they are derived from bone marrow precursors or from resident macrophages.Given that the formation of homodimers by Gal-1 and Gal-8 and pentamers by Gal-3 to generate gal-glycan lattices at the cell surface and in the extracellular space has been shown, it is possible that in PG tissue Gal-1, -3 and -8, through their binding partners, form a supramolecular structure at the surface of ECs and plump ECs, macrophages and in the extracellular space that might be mediating the transdifferentiation of macrophages into plump ECs and facilitating the migration and incorporation of these cells into the pre-existing venules, thus contributing to the formation of MVs and DVs.
Collapse
Affiliation(s)
- Enrique Arciniegas
- Institute of Biomedicine, Central University of Venezuela, Caracas, Venezuela
| | - Luz Marina Carrillo
- Institute of Biomedicine, Central University of Venezuela, Caracas, Venezuela
- Autonomus Service Institute of Biomedicine, Caracas, Venezuela
| | - Héctor Rojas
- Institute of Immunology, Central University of Venezuela, Caracas, Venezuela
| | - Jacinto Pineda
- Institute of Anatomy and Pathology, Central University of Venezuela, Caracas, Venezuela
| | - Richard Ramírez
- Autonomus Service Institute of Biomedicine, Caracas, Venezuela
| | - Oscar Reyes
- Autonomus Service Institute of Biomedicine, Caracas, Venezuela
| | - Marina Chopite
- Autonomus Service Institute of Biomedicine, Caracas, Venezuela
| | - Albani Rocheta
- Autonomus Service Institute of Biomedicine, Caracas, Venezuela
| |
Collapse
|
27
|
Docosahexaenoic acid nanoencapsulated with anti-PECAM-1 as co-therapy for atherosclerosis regression. Eur J Pharm Biopharm 2020; 159:99-107. [PMID: 33358940 DOI: 10.1016/j.ejpb.2020.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/26/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022]
Abstract
Atherosclerosis is a non-resolving inflammatory condition that underlies major cardiovascular diseases.Recent clinical trial using an anti-inflammatory drug has showna reduction of cardiovascular mortality, but increased the susceptibility to infections. For this reason, tissue target anti-inflammatory therapies can represent a better option to regress atherosclerotic plaques. Docosahexaenoic acid (DHA) is a natural omega 3 fatty acidcomponentof algae oil and acts asaprecursor of several anti-inflammatory compounds, such the specialized proresolving lipid mediators(SPMs). During the atherosclerosis process, the inflammatory condition of the endothelium leads to the higher expression of adhesion molecules, such as Endothelial Cell Adhesion Molecule Plate 1 (PECAM-1 or CD31), as part of the innate immune response. Thus, the objective of this study was to develop lipid-core nanocapsules with DHA constituting the nucleus and anti-PECAM-1 on their surface and drive this structure to the inflamed endothelium. Nanocapsules were prepared by interfacial deposition of pre-formed polymer method. Zinc-II was added to bind anti-PECAM-1 to the nanocapsule surface by forming an organometallic complex. Swelling experiment showed that the algae oil act as non-solvent for the polymer (weight constant weight for 60 days, p > 0.428) indicating an adequate material to produce kinetically stable lipid-core nanocapsules (LNC). Five formulations were synthesized: Lipid-core nanocapsules containing DHA (LNC-DHA) or containing Medium-chain triglycerides (LNC-MCT), multi-wall nanocapsules containing DHA (MLNC-DHA) or containing MCT (MLNC-MCT) and the surface-functionalized (anti-PECAM-1) metal-complex multi-wall nanocapsules containing DHA (MCMN-DHA-a1). All formulations showed homogeneous macroscopic aspects without aggregation. The mean size of the nanocapsules measured by laser diffraction did not show difference among the samples (p = 0.241). Multi-wall nanocapsules (MLNC) showed a slight increase in the mean diameter and polydispersity index (PDI) measured by DLS, lower pH and an inversion in the zeta-potential (ξP) compared to LNCs. Conjugation test for anti-PECAM-1 showed 94.80% of efficiency. The mean diameter of the formulation had slightly increased from 160 nm (LCN-DHA) and 162 nm (MLNC-DHA) to 164 nm (MCMN-DHA-a1) indicating that the surface functionalization did not induce aggregation of the nanocapsules. Biological assays showed that the MCMN-DHA-a1 were uptaken by the HUVEC cells and did not decrease their viability. The surface-functionalized (anti- PECAM-1) metal-complex multi-wall nanocapsules containing DHA (MCMN-DHA-a1) can be considered adequate for pharmaceutical approaches.
Collapse
|
28
|
Genome Wide Epistasis Study of On-Statin Cardiovascular Events with Iterative Feature Reduction and Selection. J Pers Med 2020; 10:jpm10040212. [PMID: 33171725 PMCID: PMC7712544 DOI: 10.3390/jpm10040212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/30/2020] [Accepted: 11/04/2020] [Indexed: 12/25/2022] Open
Abstract
Predicting risk for major adverse cardiovascular events (MACE) is an evidence-based practice that incorporates lifestyle, history, and other risk factors. Statins reduce risk for MACE by decreasing lipids, but it is difficult to stratify risk following initiation of a statin. Genetic risk determinants for on-statin MACE are low-effect size and impossible to generalize. Our objective was to determine high-level epistatic risk factors for on-statin MACE with GWAS-scale data. Controlled-access data for 5890 subjects taking a statin collected from Vanderbilt University Medical Center's BioVU were obtained from dbGaP. We used Random Forest Iterative Feature Reduction and Selection (RF-IFRS) to select highly informative genetic and environmental features from a GWAS-scale dataset of patients taking statin medications. Variant-pairs were distilled into overlapping networks and assembled into individual decision trees to provide an interpretable set of variants and associated risk. 1718 cases who suffered MACE and 4172 controls were obtained from dbGaP. Pathway analysis showed that variants in genes related to vasculogenesis (FDR = 0.024), angiogenesis (FDR = 0.019), and carotid artery disease (FDR = 0.034) were related to risk for on-statin MACE. We identified six gene-variant networks that predicted odds of on-statin MACE. The most elevated risk was found in a small subset of patients carrying variants in COL4A2, TMEM178B, SZT2, and TBXAS1 (OR = 4.53, p < 0.001). The RF-IFRS method is a viable method for interpreting complex "black-box" findings from machine-learning. In this study, it identified epistatic networks that could be applied to risk estimation for on-statin MACE. Further study will seek to replicate these findings in other populations.
Collapse
|
29
|
Endothelial damage and a thin intercellular fibrin network promote haemorrhage in acute promyelocytic leukaemia. EBioMedicine 2020; 60:102992. [PMID: 32949998 PMCID: PMC7501057 DOI: 10.1016/j.ebiom.2020.102992] [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: 05/26/2020] [Revised: 08/16/2020] [Accepted: 08/24/2020] [Indexed: 01/10/2023] Open
Abstract
Background The role of vascular endothelium in acute promyelocytic leukaemia (APL) remains unknown. We aimed to investigate the mechanisms by which APL cells interact with endothelial cells (ECs) and to further explore how the endothelium affects bleeding as well as therapeutic interventions. Method APL cells and an original APL cell line, NB4 cells, were used for experiments. The effects of leukaemic cells on ECs were analyzed in vitro and in vivo. Moreover, the endothelial barrier function and procoagulant activity were detected. An APL mouse model was established for in vivo studies. Findings APL cells interacted with ECs via ICAM-1 and VCAM-1 receptors to disrupt endothelial integrity. This binding activated MLCK signaling, resulting in the trans-endothelial passage of protein and red blood cells (RBCs). Combined treatment with asiatic acid or anti-adhesion receptor antibody inhibited the response of ECs to APL cells, thereby preventing APL-associated haemorrhage in vitro and in vivo. Activated ECs exhibited a procoagulant phenotype after phosphatidylserine exposure. Plasma from APL patients formed a thin fibrin network between procoagulant ECs, and this intercellular fibrin decreased the passage of albumin and RBCs. Ex vivo addition of fibrinogen further enhanced this barrier function in a dose-dependent manner. Interpretation Endothelial damage induced by leukaemic cell adherence promotes haemorrhaging in APL. Stabilization of ECs, decreasing adhesion receptor expression, and increasing fibrinogen transfusion levels may be a new therapeutic avenue to alleviate this fatal bleeding complication. Funding National Science Foundation of China (81670128, 81873433).
Collapse
|
30
|
Luo L, Xu M, Liao D, Deng J, Mei H, Hu Y. PECAM-1 protects against DIC by dampening inflammatory responses via inhibiting macrophage pyroptosis and restoring vascular barrier integrity. Transl Res 2020; 222:1-16. [PMID: 32417429 DOI: 10.1016/j.trsl.2020.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/12/2020] [Accepted: 04/07/2020] [Indexed: 12/16/2022]
Abstract
Disseminated intravascular coagulation (DIC) is a frequent complication of sepsis that affects patient outcomes due to accompanying thrombo-inflammation and microvascular permeability changes. Platelet endothelial cell adhesion molecule-1 (PECAM-1), a cellular adhesion and signaling receptor that is expressed on both hematopoietic and endothelial cells, plays an important anti-inflammatory role in acute and chronic inflammatory disease models. Little is known, however, about role and mechanism of PECAM-1 in septic DIC. Here, we investigated whether PECAM-1 might play a protective role in hindering the development of septic DIC. Plasma levels of soluble PECAM-1 were markedly elevated in septic patients that developed DIC, with a correspondingly poorer outcome. PECAM-1 knockout exhibited more severe DIC and poorer outcome in the LPS induced- and cecal ligation and puncture-induced DIC model, which could be alleviated by tissue factor inhibitor. This phenomenon seemed to be equally linked to PECAM-1 expression by both endothelial and blood cells. Furthermore, PECAM-1 was found to exert its protective effect on developing septic DIC by the following 2 distinct mechanisms: the inhibition of macrophage pyroptosis and the acceleration of the restoration of the endothelial cell barrier. Taken together, these results implicate PECAM-1 as a potentially attractive target for the development of novel therapeutics to manage and treat septic DIC.
Collapse
Affiliation(s)
- Lili Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Min Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Danying Liao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jun Deng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| |
Collapse
|
31
|
Chen CM, Lu HC, Tung YT, Chen W. Antiplatelet Therapy for Acute Respiratory Distress Syndrome. Biomedicines 2020; 8:biomedicines8070230. [PMID: 32708068 PMCID: PMC7399831 DOI: 10.3390/biomedicines8070230] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/14/2020] [Accepted: 07/18/2020] [Indexed: 12/18/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common and devastating syndrome that contributes to serious morbidities and mortality in critically ill patients. No known pharmacologic therapy is beneficial in the treatment of ARDS, and the only effective management is through a protective lung strategy. Platelets play a crucial role in the pathogenesis of ARDS, and antiplatelet therapy may be a potential medication for ARDS. In this review, we introduce the overall pathogenesis of ARDS, and then focus on platelet-related mechanisms underlying the development of ARDS, including platelet adhesion to the injured vessel wall, platelet-leukocyte-endothelium interactions, platelet-related lipid mediators, and neutrophil extracellular traps. We further summarize antiplatelet therapy, including aspirin, glycoprotein IIb/IIIa receptor antagonists, and P2Y12 inhibitors for ARDS in experimental and clinical studies and a meta-analysis. Novel aspirin-derived agents, aspirin-triggered lipoxin, and aspirin-triggered resolvin D1 are also described here. In this narrative review, we summarize the current knowledge of the role of platelets in the pathogenesis of ARDS, and the potential benefits of antiplatelet therapy for the prevention and treatment of ARDS.
Collapse
Affiliation(s)
- Chuan-Mu Chen
- Department of Life Sciences, National Chung Hsing University, 145 Xingda Road, Taichung 402, Taiwan;
- The iEGG and Animal Biotechnology Center, and the Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Hsiao-Ching Lu
- Division of Respiratory Therapy, Chia-Yi Christian Hospital, Chiayi 60002, Taiwan;
| | - Yu-Tang Tung
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei City 110, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: (Y.-T.T.); (W.C.); Tel.: +886-227361661 (Y.-T.T.); +886-5-2779365 (ext. 6172) (W.C.)
| | - Wei Chen
- Department of Life Sciences, National Chung Hsing University, 145 Xingda Road, Taichung 402, Taiwan;
- Division of Pulmonary and Critical Care Medicine, Chia-Yi Christian Hospital, Chiayi 60002, Taiwan
- Correspondence: (Y.-T.T.); (W.C.); Tel.: +886-227361661 (Y.-T.T.); +886-5-2779365 (ext. 6172) (W.C.)
| |
Collapse
|
32
|
Cheung KCP, Fanti S, Mauro C, Wang G, Nair AS, Fu H, Angeletti S, Spoto S, Fogolari M, Romano F, Aksentijevic D, Liu W, Li B, Cheng L, Jiang L, Vuononvirta J, Poobalasingam TR, Smith DM, Ciccozzi M, Solito E, Marelli-Berg FM. Preservation of microvascular barrier function requires CD31 receptor-induced metabolic reprogramming. Nat Commun 2020; 11:3595. [PMID: 32681081 PMCID: PMC7367815 DOI: 10.1038/s41467-020-17329-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/09/2020] [Indexed: 12/19/2022] Open
Abstract
Endothelial barrier (EB) breaching is a frequent event during inflammation, and it is followed by the rapid recovery of microvascular integrity. The molecular mechanisms of EB recovery are poorly understood. Triggering of MHC molecules by migrating T-cells is a minimal signal capable of inducing endothelial contraction and transient microvascular leakage. Using this model, we show that EB recovery requires a CD31 receptor-induced, robust glycolytic response sustaining junction re-annealing. Mechanistically, this response involves src-homology phosphatase activation leading to Akt-mediated nuclear exclusion of FoxO1 and concomitant β-catenin translocation to the nucleus, collectively leading to cMyc transcription. CD31 signals also sustain mitochondrial respiration, however this pathway does not contribute to junction remodeling. We further show that pathologic microvascular leakage in CD31-deficient mice can be corrected by enhancing the glycolytic flux via pharmacological Akt or AMPK activation, thus providing a molecular platform for the therapeutic control of EB response.
Collapse
Affiliation(s)
- Kenneth C P Cheung
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- School of Life Sciences, Centre for Cell & Developmental Biology and Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Silvia Fanti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Claudio Mauro
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Mindelson Way, Birmingham, B152WB, UK
| | - Guosu Wang
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Anitha S Nair
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Hongmei Fu
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Silvia Angeletti
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, Rome, Italy
| | - Silvia Spoto
- Internal Medicine Department, University campus Bio-Medico of Rome, Rome, Italy
| | - Marta Fogolari
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, Rome, Italy
| | - Francesco Romano
- Unit of Clinical Laboratory Science, University Campus Bio-Medico of Rome, Rome, Italy
| | - Dunja Aksentijevic
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Weiwei Liu
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, 510060, People's Republic of China
| | - Baiying Li
- School of Life Sciences, Centre for Cell & Developmental Biology and Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lixin Cheng
- School of Life Sciences, Centre for Cell & Developmental Biology and Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Liwen Jiang
- School of Life Sciences, Centre for Cell & Developmental Biology and Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Juho Vuononvirta
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Thanushiyan R Poobalasingam
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - David M Smith
- AstraZeneca R&D, Cambridge Science Park, Milton Road, Cambridge, CB4 0WG, UK
| | - Massimo Ciccozzi
- Unit of Medical Statistic and Molecular Epidemiology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Egle Solito
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universita degli studi di Napoli "Federico II", 80131, Naples, Italy
| | - Federica M Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
- Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, Charterhouse Square, London, UK.
| |
Collapse
|
33
|
Wright WS, Eshaq RS, Lee M, Kaur G, Harris NR. Retinal Physiology and Circulation: Effect of Diabetes. Compr Physiol 2020; 10:933-974. [PMID: 32941691 PMCID: PMC10088460 DOI: 10.1002/cphy.c190021] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In this article, we present a discussion of diabetes and its complications, including the macrovascular and microvascular effects, with the latter of consequence to the retina. We will discuss the anatomy and physiology of the retina, including aspects of metabolism and mechanisms of oxygenation, with the latter accomplished via a combination of the retinal and choroidal blood circulations. Both of these vasculatures are altered in diabetes, with the retinal circulation intimately involved in the pathology of diabetic retinopathy. The later stages of diabetic retinopathy involve poorly controlled angiogenesis that is of great concern, but in our discussion, we will focus more on several alterations in the retinal circulation occurring earlier in the progression of disease, including reductions in blood flow and a possible redistribution of perfusion that may leave some areas of the retina ischemic and hypoxic. Finally, we include in this article a more recent area of investigation regarding the diabetic retinal vasculature, that is, the alterations to the endothelial surface layer that normally plays a vital role in maintaining physiological functions. © 2020 American Physiological Society. Compr Physiol 10:933-974, 2020.
Collapse
Affiliation(s)
- William S Wright
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, South Carolina, USA
| | - Randa S Eshaq
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Minsup Lee
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Gaganpreet Kaur
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Norman R Harris
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
34
|
Duong CN, Vestweber D. Mechanisms Ensuring Endothelial Junction Integrity Beyond VE-Cadherin. Front Physiol 2020; 11:519. [PMID: 32670077 PMCID: PMC7326147 DOI: 10.3389/fphys.2020.00519] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/27/2020] [Indexed: 12/30/2022] Open
Abstract
Endothelial junctions provide blood and lymph vessel integrity and are essential for the formation of a vascular system. They control the extravasation of solutes, leukocytes and metastatic cells from blood vessels and the uptake of fluid and leukocytes into the lymphatic vascular system. A multitude of adhesion molecules mediate and control the integrity and permeability of endothelial junctions. VE-cadherin is arguably the most important adhesion molecule for the formation of vascular structures, and the stability of their junctions. Interestingly, despite this prominence, its elimination from junctions in the adult organism has different consequences in the vasculature of different organs, both for blood and lymph vessels. In addition, even in tissues where the lack of VE-cadherin leads to strong plasma leaks from venules, the physical integrity of endothelial junctions is preserved. Obviously, other adhesion molecules can compensate for a loss of VE-cadherin and this review will discuss which other adhesive mechanisms contribute to the stability and regulation of endothelial junctions and cooperate with VE-cadherin in intact vessels. In addition to adhesion molecules, endothelial receptors will be discussed, which stimulate signaling processes that provide junction stability by modulating the actomyosin system, which reinforces tension of circumferential actin and dampens pulling forces of radial stress fibers. Finally, we will highlight most recent reports about the formation and control of the specialized button-like junctions of initial lymphatics, which represent the entry sites for fluid and cells into the lymphatic vascular system.
Collapse
Affiliation(s)
| | - Dietmar Vestweber
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
35
|
Xie X, Wang F, Zhu L, Yang H, Pan D, Liu Y, Qu X, Gu Y, Li X, Chen S. Low shear stress induces endothelial cell apoptosis and monocyte adhesion by upregulating PECAM‑1 expression. Mol Med Rep 2020; 21:2580-2588. [PMID: 32323830 PMCID: PMC7185273 DOI: 10.3892/mmr.2020.11060] [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] [Received: 10/13/2019] [Accepted: 03/19/2020] [Indexed: 01/02/2023] Open
Abstract
Low shear stress serves an important role in the initiation and progression of atherosclerotic lesions, with an impact on progression, but its detailed mechanisms are .not yet fully known. The present study aimed to investigate endothelial cell (EC) apoptosis, as well as monocyte adhesion induced by low shear stress and the potential underlying mechanisms. The expression of platelet endothelial cell adhesion molecule-1 (PECAM-1) was demonstrated to be enhanced in human umbilical vascular ECs with a trend that was associated with time when stimulated by low shear stress compared with unstimulated cells. EC apoptosis was increased under low shear stress compared with unstimulated cells, and knockdown of PECAM-1 inhibited this process. Furthermore, downregulation of PECAM-1 reduced monocyte adhesion induced by low shear stress compared with that in the negative control cells. Mechanistically, PECAM-1 small interfering RNA transfection increased Akt and forkhead box O1 phosphorylation under low shear stress conditions compared with that in the negative control cells. Collectively, the findings of the present study revealed that low shear stress induced EC apoptosis and monocyte adhesion by upregulating PECAM-1 expression, which suggested that PECAM-1 may be a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Xiangrong Xie
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Feng Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Linlin Zhu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Hongfeng Yang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Daorong Pan
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Yan Liu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Xinliang Qu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Yue Gu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Xiaobo Li
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Shaoliang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| |
Collapse
|
36
|
Harris NR, Leskova W, Kaur G, Eshaq RS, Carter PR. Blood flow distribution and the endothelial surface layer in the diabetic retina. Biorheology 2020; 56:181-189. [PMID: 30958328 PMCID: PMC10082436 DOI: 10.3233/bir-180200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetic retinopathy is known as a microvascular complication of hyperglycemia, with a breakdown of the blood-retinal barrier, loss of pericytes, formation of microhemorrhages, early decreases in perfusion and areas of ischemia, with the latter speculated to induce the eventual proliferative, angiogenic phase of the disease. Our animal models of diabetic retinopathy demonstrate similar decreases in retinal blood flow as seen in the early stages of diabetes in humans. Our studies also show an alteration in the retinal distribution of red blood cells, with the deep capillary layer receiving a reduced fraction, and with flow being diverted more towards the superficial vascular layer. Normal red blood cell distribution is dependent on the presence of the endothelial surface layer, specifically the glycocalyx, which has been reported to be partially lost in the diabetic retina of both humans and animals. This review addresses these two phenomena in diabetes: altered perfusion patterns and loss of the glycocalyx, with a possible connection between the two.
Collapse
Affiliation(s)
- Norman R Harris
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Wendy Leskova
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Gaganpreet Kaur
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Randa S Eshaq
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Patsy R Carter
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
37
|
Duong CN, Nottebaum AF, Butz S, Volkery S, Zeuschner D, Stehling M, Vestweber D. Interference With ESAM (Endothelial Cell-Selective Adhesion Molecule) Plus Vascular Endothelial-Cadherin Causes Immediate Lethality and Lung-Specific Blood Coagulation. Arterioscler Thromb Vasc Biol 2020; 40:378-393. [DOI: 10.1161/atvbaha.119.313545] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Objective:
Vascular endothelial (VE)-cadherin is of dominant importance for the formation and stability of endothelial junctions, yet induced gene inactivation enhances vascular permeability in the lung but does not cause junction rupture. This study aims at identifying the junctional adhesion molecule, which is responsible for preventing endothelial junction rupture in the pulmonary vasculature in the absence of VE-cadherin.
Approach and Results:
We have compared the relevance of ESAM (endothelial cell-selective adhesion molecule), JAM (junctional adhesion molecule)-A, PECAM (platelet endothelial cell adhesion molecule)-1, and VE-cadherin for vascular barrier integrity in various mouse tissues. Gene inactivation of ESAM enhanced vascular permeability in the lung but not in the heart, skin, and brain. In contrast, deletion of JAM-A or PECAM-1 did not affect barrier integrity in any of these organs. Blocking VE-cadherin with antibodies caused lethality in ESAM
−/−
mice within 30 minutes but had no such effect in JAM-A
−/−
, PECAM-1
−/−
or wild-type mice. Likewise, induced gene inactivation of VE-cadherin caused rapid lethality only in the absence of ESAM. Ultrastructural analysis revealed that only combined interference with VE-cadherin and ESAM disrupted endothelial junctions and caused massive blood coagulation in the lung. Mechanistically, we could exclude a role of platelet ESAM in coagulation, changes in the expression of other junctional proteins or a contribution of cytoplasmic signaling domains of ESAM.
Conclusions:
Despite well-documented roles of JAM-A and PECAM-1 for the regulation of endothelial junctions, only for ESAM, we detected an essential role for endothelial barrier integrity in a tissue-specific way. In addition, we found that it is ESAM which prevents endothelial junction rupture in the lung when VE-cadherin is absent.
Collapse
Affiliation(s)
- Cao Nguyen Duong
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Astrid F. Nottebaum
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Stefan Butz
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Stefan Volkery
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Dagmar Zeuschner
- Electron Microscopy and Flow Cytometry Unit (D.Z., M.S.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Martin Stehling
- Electron Microscopy and Flow Cytometry Unit (D.Z., M.S.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Dietmar Vestweber
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
38
|
Li M, Alfieri CM, Morello W, Cellesi F, Armelloni S, Mattinzoli D, Montini G, Messa P. Assessment of increased glomerular permeability associated with recurrent focal segmental glomerulosclerosis using an in vitro model of the glomerular filtration barrier. J Nephrol 2019; 33:747-755. [PMID: 31853790 DOI: 10.1007/s40620-019-00683-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/02/2019] [Indexed: 11/25/2022]
Abstract
The presence of circulating permeability factors (cPFs) has been hypothesized to be associated with recurrence of focal segmental glomerulosclerosis (rFSGS) in renal allografts. The available methods to detect cPFs are complex, not easily repeatable and inappropriate to represent the anatomical characteristics of the three-layer glomerular filtration barrier (GFB). Here we describe a novel method which measures the permeability to bovine serum albumin (BSA) through a three-layer device (3LD). The 3 layers comprise: (1) conditionally immortalized human podocytes (HCiPodo), (2) collagen type IV coated porous membrane and (3) human glomerular endothelial cells (HCiGEnC). Using this method, we found that sera from all rFSGS patients increased albumin permeability, while sera from non recurrent (nrFSGS) and genetic (gFSGS) forms of FSGS did not. The mechanisms underlying the increase of albumin permeability are probably due to endothelial cell damage as an initial event, which was demonstrated by the decrease of Platelet endothelial cell adhesion molecule (PECAM-1 or CD31), while the podocytes' expressions of synaptopodin and podocin were normal. Furthermore, we also found that the plasmapheretic treatment (PPT) eliminated the effect of increasing BSA permeability in sera from rFSGS patients. These preliminary data suggest that our in vitro GFB model could not only be useful in predicting the recurrence of FSGS after renal transplantation (RTx), but also be a valuable in vitro model to study podocyte and endothelial cell biology.
Collapse
Affiliation(s)
- Min Li
- IRCCS Ospedale Maggiore Policlinico, Renal Research Laboratory, Foundation Ca' Granda, Milan, Italy
| | - Carlo Maria Alfieri
- Unit of Adult Nephrology, Dialysis and Renal Transplant, Department of Medicine, Foundation Ca' Granda IRCCS Ospedale Maggiore Policlinico, Via Commenda 15, 20122, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli studi di Milano, Milan, Italy
| | - William Morello
- Pediatric Nephrology, Dialysis and Transplant Unit, Foundation IRCCS Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Cellesi
- Politecnico di Milano, Dipartimento di Chimica, Materiali ed Ingegneria Chimica "G. Natta", Milan, Italy
| | - Silvia Armelloni
- IRCCS Ospedale Maggiore Policlinico, Renal Research Laboratory, Foundation Ca' Granda, Milan, Italy
| | - Deborah Mattinzoli
- IRCCS Ospedale Maggiore Policlinico, Renal Research Laboratory, Foundation Ca' Granda, Milan, Italy
| | - Giovanni Montini
- Pediatric Nephrology, Dialysis and Transplant Unit, Foundation IRCCS Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli studi di Milano, Milan, Italy
| | - Piergiorgio Messa
- Unit of Adult Nephrology, Dialysis and Renal Transplant, Department of Medicine, Foundation Ca' Granda IRCCS Ospedale Maggiore Policlinico, Via Commenda 15, 20122, Milan, Italy.
- Department of Clinical Sciences and Community Health, Università degli studi di Milano, Milan, Italy.
| |
Collapse
|
39
|
Khegay II. Noncanonical effects of vasopressin in angiogenesis. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj19.527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022] Open
Abstract
The molecular action of vasopressin depends on the localization of hormonal receptors. The basic physiological effects of vasopressin are manifested in the blood vasculature, renal inner medulla and brain. To date, new information concerning the tissue-specific spreading of vasopressin receptors has been accumulated, and it needs to be summarized. Platelets and endotheliocytes expressing V1a and V2 receptor types, respectively, are related to less investigated targets of the hormone. Vasopressin induces the initial reversible stage of platelet activation, required for interaction with intercellular matrix proteins. Platelet adhesion on endothelium activates cellular secretion of growth factors and enzymes for intercellular matrix glucosamine metabolism. Platelet hyaluronidase HYAL2 hydrolyses high-molecular hyaluronic acid to shorter fragments. Unlike intact hyaluronic acid with a molecular weight of several megadaltons, generally showing distinctive antiangiogenic properties, intermediate fractions of hyaluronan hydrolysis in a range from 2.5 to 200 kilodaltons have a stimulating effect on angiogenesis. Intercellular contacts between platelets and endotheliocytes are stabilized due to adhesive transmembrane glycoprotein PECAM-1 interaction. Resulting PECAM-1 heterodimers acquire conformation with high affinity to integrins αvβ3. Integrin activation forms contact links between endothelium and fibrillar proteins. Activated endotheliocytes secrete von Willebrand factor and P-selectin. These proteins are accumulated in Weibel–Palade bodies. Vasopressin stimulates cAMP-dependent ACAP-regulated exocytosis of Weibel–Palade bodies. von Willebrand factor possesses adhesive properties and additionally accelerates interaction of cells with the intercellular matrix. Adhesion on fibrillar collagen and membrane glycoproteins in cooperation with effects of PECAM-1–αvβ3 integrin complexes fixes cell aggregates in the surrounding interstitium and promotes proliferating endotheliocyte migration in according to the direction of local growth factor gradients during angiogenesis. Neurohormonal regulation of platelet and endotheliocyte secretory activity functionally link proliferation and migration of endotheliocytes during angiogenesis and integrate it according to the adaptive capacity of the entire organism.
Collapse
|
40
|
Eshaq RS, Harris NR. Loss of Platelet Endothelial Cell Adhesion Molecule-1 (PECAM-1) in the Diabetic Retina: Role of Matrix Metalloproteinases. Invest Ophthalmol Vis Sci 2019; 60:748-760. [PMID: 30793207 PMCID: PMC6385619 DOI: 10.1167/iovs.18-25068] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To test the hypothesis that high glucose and matrix metalloproteinases (MMPs) contribute to the diabetes-induced loss of platelet endothelial cell adhesion molecule-1 (PECAM-1) in the retinal microvasculature. Methods PECAM-1 and MMP protein, activity, and interactions with PECAM-1 were assessed using western blotting, zymography, immunofluorescence, or coimmunoprecipitation assays. These assays were conducted using primary rat retinal microvascular endothelial cells (RRMECs) grown either in normal glucose (5 mM) or high glucose (25 mM) conditions and using retinas collected from streptozotocin-induced diabetic or control rats. The broad-spectrum MMP inhibitor GM6001 was administered in vivo and in vitro to ascertain the role of MMPs in the hyperglycemia-induced loss of PECAM-1. Results A dramatic decrease in PECAM-1 (western blotting, immunofluorescence) was observed in both the diabetic retina and in hyperglycemic RRMECs. The decrease in PECAM-1 was accompanied by a significant increase in the presence and activity of matrix metalloproteinase-2 (MMP-2) (but not matrix metalloproteinase-9 [MMP-9]) in the diabetic plasma (P < 0.05) and in hyperglycemic RRMECs (P < 0.05). Moreover, RRMEC PECAM-1 significantly decreased when treated with plasma collected from diabetic rats. Several MMP-2 cleavage sites on PECAM-1 were identified using in silico analysis. Moreover, PECAM-1/MMP-2 interactions were confirmed using coimmunoprecipitation. PECAM-1 was significantly decreased in RRMECs treated with MMP-2 (P < 0.05), but became comparable to controls with the MMP inhibitor GM6001 in both the diabetic retina and hyperglycemic RRMECs. Conclusions These results indicate a possible role of MMP-2 in hyperglycemia-induced PECAM-1 loss in retinal endothelial cells.
Collapse
Affiliation(s)
- Randa S Eshaq
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States
| | - Norman R Harris
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States
| |
Collapse
|
41
|
Wimmer I, Tietz S, Nishihara H, Deutsch U, Sallusto F, Gosselet F, Lyck R, Muller WA, Lassmann H, Engelhardt B. PECAM-1 Stabilizes Blood-Brain Barrier Integrity and Favors Paracellular T-Cell Diapedesis Across the Blood-Brain Barrier During Neuroinflammation. Front Immunol 2019; 10:711. [PMID: 31024547 PMCID: PMC6460670 DOI: 10.3389/fimmu.2019.00711] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 03/15/2019] [Indexed: 01/13/2023] Open
Abstract
Breakdown of the blood-brain barrier (BBB) and increased immune cell trafficking into the central nervous system (CNS) are hallmarks of the pathogenesis of multiple sclerosis (MS). Platelet endothelial cell adhesion molecule-1 (PECAM-1; CD31) is expressed on cells of the vascular compartment and regulates vascular integrity and immune cell trafficking. Involvement of PECAM-1 in MS pathogenesis has been suggested by the detection of increased levels of soluble PECAM-1 (sPECAM-1) in the serum and CSF of MS patients. Here, we report profound upregulation of cell-bound PECAM-1 in initial (pre-phagocytic) white matter as well as active cortical gray matter MS lesions. Using a human in vitro BBB model we observed that PECAM-1 is not essential for the transmigration of human CD4+ T-cell subsets (Th1, Th1*, Th2, and Th17) across the BBB. Employing an additional in vitro BBB model based on primary mouse brain microvascular endothelial cells (pMBMECs) we show that the lack of endothelial PECAM-1 impairs BBB properties as shown by reduced transendothelial electrical resistance (TEER) and increases permeability for small molecular tracers. Investigating T-cell migration across the BBB under physiological flow by in vitro live cell imaging revealed that absence of PECAM-1 in pMBMECs did not influence arrest, polarization, and crawling of effector/memory CD4+ T cells on the pMBMECs. Absence of endothelial PECAM-1 also did not affect the number of T cells able to cross the pMBMEC monolayer under flow, but surprisingly favored transcellular over paracellular T-cell diapedesis. Taken together, our data demonstrate that PECAM-1 is critically involved in regulating BBB permeability and although not required for T-cell diapedesis itself, its presence or absence influences the cellular route of T-cell diapedesis across the BBB. Upregulated expression of cell-bound PECAM-1 in human MS lesions may thus reflect vascular repair mechanisms aiming to restore BBB integrity and paracellular T-cell migration across the BBB as it occurs during CNS immune surveillance.
Collapse
Affiliation(s)
- Isabella Wimmer
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Silvia Tietz
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, ETH Zürich,, Zurich, Switzerland
| | - Fabien Gosselet
- Blood-Brain Barrier Laboratory, Université d'Artois, Lens, France
| | - Ruth Lyck
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - William A. Muller
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
42
|
Kuriri FA, O'Malley CJ, Jackson DE. Molecular mechanisms of immunoreceptors in platelets. Thromb Res 2019; 176:108-114. [DOI: 10.1016/j.thromres.2019.01.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/20/2019] [Accepted: 01/28/2019] [Indexed: 01/05/2023]
|
43
|
Li Y, Wang S, Zhang D, Xu X, Yu B, Zhang Y. The association of functional polymorphisms in genes expressed in endothelial cells and smooth muscle cells with the myocardial infarction. Hum Genomics 2019; 13:5. [PMID: 30678728 PMCID: PMC6345039 DOI: 10.1186/s40246-018-0189-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 12/27/2018] [Indexed: 01/05/2024] Open
Abstract
Background The association of platelet endothelial cell adhesion molecule 1 (PECAM1), hypoxia-inducible factor 1 subunit alpha (HIF1A), and KIAA1462 in myocardial infarction (MI) was investigated. The study included 401 Han Chinese MI patients and 409 controls. Three tag single-nucleotide polymorphisms (SNPs)—PECAM1 rs1867624, HIF1A rs2057482, and KIAA1462 rs3739998—were selected. SNP genotyping was performed by an improved multiplex ligation detection reaction assay. A systematic review and meta-analysis of studies including 3314 cases and 2687 controls on the association of 5 HIF1A SNPs and the overall risk of MI or coronary artery disease (CAD) was performed. Results The rs1867624 variants were associated with high TG concentrations (p = 0.040) and the rs2057482 variants were associated with decreased HDL-C in MI patients compared with the control group (p = 0.003). Rs2057482 SNP interacted with age to influence TC levels. The SNP of rs3739998 interacted with sex and hypertension to modulate CRE and TG levels, respectively (p < 3.04E-5-0.002). No association between the three SNPs and susceptibility to MI was found (p > 0.05 for all). In the meta-analysis of HIF1A, the rs11549465 C > T and rs10873142 T > C polymorphisms, but not rs2057482, rs11549467, and rs41508050, were correlated with overall MI or CAD risk. Conclusions Taken together, this study provides additional evidence that genetic variation of the PECAM1 rs1867624 and HIF1A rs2057482 can mediate lipid levels in MI patients. Electronic supplementary material The online version of this article (10.1186/s40246-018-0189-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yilan Li
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Shipeng Wang
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Dandan Zhang
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Xueming Xu
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Bo Yu
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Yao Zhang
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China. .,Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
44
|
Stolwijk JA, Wegener J. Impedance-Based Assays Along the Life Span of Adherent Mammalian Cells In Vitro: From Initial Adhesion to Cell Death. BIOANALYTICAL REVIEWS 2019. [DOI: 10.1007/11663_2019_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
45
|
Shi P, Cao Y, Gao J, Fu B, Ren J, Ba L, Song C, Qi H, Huang W, Guan X, Sun H. Allicin improves the function of cardiac microvascular endothelial cells by increasing PECAM-1 in rats with cardiac hypertrophy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 51:241-254. [PMID: 30466623 DOI: 10.1016/j.phymed.2018.10.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 10/13/2018] [Accepted: 10/18/2018] [Indexed: 06/09/2023]
Abstract
OBJECTIVE Cardiac microvascular damage is significantly associated with the development of cardiac hypertrophy (CH). Researchers found that allicin could inhibit CH, but the relationship between cardiac microvessel and the inhibition of allicin on CH has not been reported. We aimed to investigate the effect of allicin on the function of cardiac microvascular endothelial cells (CMECs) in CH rat. MATERIALS AND METHODS The hemodynamic parameters were measured by BL-420F biological function experimental system and the indicators of the ventricular structure and function were measured by echocardiographic system. MTT assay was performed to assess the cell viability. Nitrite detection was performed to detect nitric oxide content. The morphology and molecular characteristics were detected by electron micrographs, immunofluorescence, quantitative real-time polymerase chain reaction (qRT-PCR), western blot. Wound healing experiment, analysis of tube formation and shear adaptation were performed to assess CMECs migration ability, angiogenesis and shear-responsiveness respectively. RESULT Our findings have identified that microvascular density was decreased by observing the expression of platelet endothelial cell adhesion molecule-1 (PECAM-1) in CH rats. Interestingly, allicin improved the distribution and expression of PECAM-1. Meanwhile, allicin enhanced the migration and angiogenesis ability of CMECs, activated PECAM-1-PI3K-AKT-eNOS signaling pathway, however, the role of allicin was disappear after PECAM-1 was silenced. Allicin decreased the expression of caspase-3 and receptor interacting protein 3 (RIP3), inhibited necroptosis, and increased the levels of Angiopoietin-2 (Ang-2) and platelet-derived growth factor receptor-β (PDGFR-β). Under 10 dyn/cm2 condition, allicin advanced the modification ability of CMECs's shear-adaptation by activating PECAM-1. CONCLUSION Allicin provided cardioprotection for CH rats by improving the function of CMECs through increasing the expression of PECAM-1.
Collapse
Affiliation(s)
- Pilong Shi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Yonggang Cao
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, China
| | - Jingquan Gao
- Department of Nursing, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Bowen Fu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Jing Ren
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Lina Ba
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Chao Song
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Hanping Qi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Wei Huang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Xueying Guan
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China.
| |
Collapse
|
46
|
Villar J, Zhang H, Slutsky AS. Lung Repair and Regeneration in ARDS: Role of PECAM1 and Wnt Signaling. Chest 2018; 155:587-594. [PMID: 30392791 DOI: 10.1016/j.chest.2018.10.022] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 10/18/2018] [Accepted: 10/18/2018] [Indexed: 01/08/2023] Open
Abstract
ARDS is an acute inflammatory pulmonary process triggered by severe pulmonary and systemic insults to the alveolar-capillary membrane. This causes increased vascular permeability and the development of interstitial and alveolar protein-rich edema, leading to acute hypoxemic respiratory failure. Supportive treatment includes the use of lung-protective ventilatory strategies that decrease the work of breathing, can improve oxygenation, and minimize ventilator-induced lung injury. Despite substantial advances in supportive measures, there are no specific pharmacologic treatments for ARDS, and the overall hospital mortality rate remains about 40% in most series. The pathophysiology of ARDS involves interactions among multiple mechanisms, including immune cell infiltration, cytokine storm, alveolar-capillary barrier disruption, cell apoptosis, and the development of fibrosis. Here we review some new developments in the molecular basis of lung injury, with a focus on possible novel pharmacologic interventions aimed at improving the outcomes of patients with ARDS. Our focus is on platelet-endothelial cell adhesion molecule-1, which contributes to the maintenance and restoration of vascular integrity following barrier disruption. We also highlight the wingless-related integration site signaling pathway, which appears to be a central mechanism for lung healing as well as for fibrotic development.
Collapse
Affiliation(s)
- Jesús Villar
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain; Multidisciplinary Organ Dysfunction Evaluation Research Network, Research Unit, Hospital Universitario Dr Negrin, Las Palmas de Gran Canaria, Spain; Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
| | - Haibo Zhang
- Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada; Department of Anesthesia and Department of Physiology, University of Toronto, Toronto, Canada
| | - Arthur S Slutsky
- Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
47
|
Vieira RF, Breithaupt-Faloppa AC, Matsubara BC, Rodrigues G, Sanches MP, Armstrong-Jr R, Ferreira SG, Correia CDJ, Moreira LFP, Sannomiya P. 17β-Estradiol protects against lung injuries after brain death in male rats. J Heart Lung Transplant 2018; 37:1381-1387. [PMID: 30139547 DOI: 10.1016/j.healun.2018.06.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/09/2018] [Accepted: 06/21/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Brain death elicits microvascular dysfunction and inflammation, and thereby compromises lung viability for transplantation. As 17β-estradiol was shown to be anti-inflammatory and vascular protective, we investigated its effects on lung injury after brain death in male rats. METHODS Wistar rats were assigned to: sham-operation by trepanation only (SH, n = 7); brain death (BD, n = 7); administration of 17β-estradiol (280 μg/kg, iv) at 60 minutes after brain death (BD-E2, n = 7). Experiments were performed 180 minutes thereafter. Histopathological changes in the lung were evaluated by histomorphometry. Gene expression of inducible nitric oxide synthase (iNOS), endothelial nitric oxide synthase (eNOS), and endothelin-1 was measured by real-time polymerase chain reaction. Protein expression of NO synthases, endothelin-1, platelet endothelial cell adhesion molecule-1 (PECAM-1), vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), BCL-2, and caspase 3 was assessed by immunohistochemistry. Cytokines were quantified by enzyme-linked immunosorbent assay. RESULTS Treatment with 17β-estradiol after brain death decreased lung edema and hemorrhage (p < 0.0001), and serum levels of cytokine-induced neutrophil chemoattractant-1 (CINC-1; p = 0.0020). iNOS (p < 0.0001) and VCAM-1 (p < 0.0001) also diminished at protein levels, while eNOS accumulated (p = 0.0002). However, gene expression of iNOS, eNOS, and endothelin-1 was comparable among groups, as was protein expression of endothelin-1, ICAM-1, BCL-2, and caspase 3. CONCLUSIONS 17β-Estradiol effectively reduces lung injury in brain-dead rats mainly due to its ability to regulate NO synthases. Thus, the drug may improve lung viability for transplantation.
Collapse
Affiliation(s)
- Roberta Figueiredo Vieira
- Laboratório Cirúrgico de Pesquisa Cardiovascular, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Cristina Breithaupt-Faloppa
- Laboratório Cirúrgico de Pesquisa Cardiovascular, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Bruno Carvalho Matsubara
- Laboratório Cirúrgico de Pesquisa Cardiovascular, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Geovana Rodrigues
- Laboratório Cirúrgico de Pesquisa Cardiovascular, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Marcelo Petrof Sanches
- Laboratório Cirúrgico de Pesquisa Cardiovascular, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Roberto Armstrong-Jr
- Laboratório Cirúrgico de Pesquisa Cardiovascular, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Sueli Gomes Ferreira
- Laboratório Cirúrgico de Pesquisa Cardiovascular, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Cristiano de Jesus Correia
- Laboratório Cirúrgico de Pesquisa Cardiovascular, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Felipe P Moreira
- Laboratório Cirúrgico de Pesquisa Cardiovascular, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Paulina Sannomiya
- Laboratório Cirúrgico de Pesquisa Cardiovascular, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
48
|
Fava C, Montagnana M. Atherosclerosis Is an Inflammatory Disease which Lacks a Common Anti-inflammatory Therapy: How Human Genetics Can Help to This Issue. A Narrative Review. Front Pharmacol 2018; 9:55. [PMID: 29467655 PMCID: PMC5808208 DOI: 10.3389/fphar.2018.00055] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/15/2018] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis is a multifactorial disease triggered and sustained by different risk factors such as dyslipidemia, arterial hypertension, diabetes mellitus, smoke, etc. Since a couple of decades, a pivotal role for inflammation in its pathogenesis has been recognized and proved at molecular levels, and already described in many animal models. Despite all this knowledge, due to the complexity of the specific inflammatory process subtending atherosclerosis and to the fact that inflammation is also a protective response against microorganisms, no anti-inflammatory therapy has been rendered available in the therapeutic armamentarium against atherosclerosis and vascular events till 2017 when canakinumab in the first ad-hoc randomized clinical trial (RCT) proved for the first time that targeting specifically inflammation lowers cardiovascular (CV) events. From the genetic side, in the 90's and early 2000, several genetic markers in inflammatory pathway have been explored searching for an association with athero-thrombosis which gave seldom consistent results. Then, in the genomic era, plenty of genetic markers covering most of the genome have been analyzed at once without a priori information. The results coming from genome wide association studies (GWAS) have pinpointed some loci closed to inflammatory molecules consistently associated with atherosclerosis and CV consequences revamping the strict link between inflammation and atherosclerosis and suggesting some tailored target therapy. Whole-exome and whole-genome sequencing will come soon showing new and old loci associated with atherosclerosis suggesting new molecular targets or underlying which inflammatory pathway could be most attractive to target for blocking atherosclerosis even in its early stages.
Collapse
Affiliation(s)
- Cristiano Fava
- General Medicine and Hypertension Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Martina Montagnana
- Clinical Biochemistry Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
49
|
Vascular endothelial effects of collaborative binding to platelet/endothelial cell adhesion molecule-1 (PECAM-1). Sci Rep 2018; 8:1510. [PMID: 29367646 PMCID: PMC5784113 DOI: 10.1038/s41598-018-20027-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/11/2018] [Indexed: 11/23/2022] Open
Abstract
Targeting drugs to endothelial cells has shown the ability to improve outcomes in animal models of inflammatory, ischemic and thrombotic diseases. Previous studies have revealed that certain pairs of ligands (antibodies and antibody fragments) specific for adjacent, but distinct, epitopes on PECAM-1 enhance each other’s binding, a phenomenon dubbed Collaborative Enhancement of Paired Affinity Ligands, or CEPAL. This discovery has been leveraged to enable simultaneous delivery of multiple therapeutics to the vascular endothelium. Given the known role of PECAM-1 in promoting endothelial quiescence and cell junction integrity, we sought here to determine if CEPAL might induce unintended vascular effects. Using a combination of in vitro and in vivo techniques and employing human and mouse endothelial cells under physiologic and pathologic conditions, we found only modest or non-significant effects in response to antibodies to PECAM-1, whether given solo or in pairs. In contrast, these methods detected significant elevation of endothelial permeability, pro-inflammatory vascular activation, and systemic cytokine release following antibody binding to the related endothelial junction protein, VE-Cadherin. These studies support the notion that PECAM-1-targeted CEPAL provides relatively well-tolerated endothelial drug delivery. Additionally, the analysis herein creates a template to evaluate potential toxicities of vascular-targeted nanoparticles and protein therapeutics.
Collapse
|
50
|
Liao D, Mei H, Hu Y, Newman DK, Newman PJ. CRISPR-mediated deletion of the PECAM-1 cytoplasmic domain increases receptor lateral mobility and strengthens endothelial cell junctional integrity. Life Sci 2018; 193:186-193. [PMID: 29122551 DOI: 10.1016/j.lfs.2017.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/24/2017] [Accepted: 11/04/2017] [Indexed: 10/18/2022]
Abstract
AIMS PECAM-1 is an abundant endothelial cell surface receptor that becomes highly enriched at endothelial cell-cell junctions, where it functions to mediate leukocyte transendothelial migration, sense changes in shear and flow, and maintain the vascular permeability barrier. Homophilic interactions mediated by the PECAM-1 extracellular domain are known to be required for PECAM-1 to perform these functions; however, much less is understood about the role of its cytoplasmic domain in these processes. MAIN METHODS CRISPR/Cas9 gene editing technology was employed to generate human endothelial cell lines that either lack PECAM-1 entirely, or express mutated PECAM-1 missing the majority of its cytoplasmic domain (∆CD-PECAM-1). The endothelial barrier function was evaluated by Electric Cell-substrate Impedance Sensing, and molecular mobility was assessed by fluorescence recovery after photobleaching. KEY FINDINGS We found that ∆CD-PECAM-1 concentrates normally at endothelial cell junctions, but has the unexpected property of conferring increased baseline barrier resistance, as well as a more rapid rate of recovery of vascular integrity following thrombin-induced disruption of the endothelial barrier. Fluorescence recovery after photobleaching analysis revealed that ∆CD-PECAM-1 exhibits increased mobility within the plane of the plasma membrane, thus allowing it to redistribute more rapidly back to endothelial cell-cell borders to reform the vascular permeability barrier. SIGNIFICANCE The PECAM-1 cytoplasmic domain plays a novel role in regulating the rate and extent of vascular permeability following thrombotic or inflammatory challenge.
Collapse
Affiliation(s)
- Danying Liao
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, United States; Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Debra K Newman
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, United States; Department of Pharmacology, Medical College of Wisconsin, Milwaukee, United States; Department of Microbiology, Medical College of Wisconsin, Milwaukee, United States; The Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
| | - Peter J Newman
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, United States; Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Department of Pharmacology, Medical College of Wisconsin, Milwaukee, United States; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, United States; The Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States.
| |
Collapse
|