1
|
Brandon KD, Frank WE, Stroka KM. Junctions at the crossroads: the impact of mechanical cues on endothelial cell-cell junction conformations and vascular permeability. Am J Physiol Cell Physiol 2024; 327:C1073-C1086. [PMID: 39129490 PMCID: PMC11481987 DOI: 10.1152/ajpcell.00605.2023] [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: 11/08/2023] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
Cells depend on precisely regulating barrier function within the vasculature to maintain physiological stability and facilitate essential substance transport. Endothelial cells achieve this through specialized adherens and tight junction protein complexes, which govern paracellular permeability across vascular beds. Adherens junctions, anchored by vascular endothelial (VE)-cadherin and associated catenins to the actin cytoskeleton, mediate homophilic adhesion crucial for barrier integrity. In contrast, tight junctions composed of occludin, claudin, and junctional adhesion molecule A interact with Zonula Occludens proteins, reinforcing intercellular connections essential for barrier selectivity. Endothelial cell-cell junctions exhibit dynamic conformations during development, maturation, and remodeling, regulated by local biochemical and mechanical cues. These structural adaptations play pivotal roles in disease contexts such as chronic inflammation, where junctional remodeling contributes to increased vascular permeability observed in conditions from cancer to cardiovascular diseases. Conversely, the brain microvasculature's specialized junctional arrangements pose challenges for therapeutic drug delivery due to their unique molecular compositions and tight organization. This commentary explores the molecular mechanisms underlying endothelial cell-cell junction conformations and their implications for vascular permeability. By highlighting recent advances in quantifying junctional changes and understanding mechanotransduction pathways, we elucidate how physical forces from cellular contacts and hemodynamic flow influence junctional dynamics.
Collapse
Affiliation(s)
- Ken D Brandon
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States
| | - William E Frank
- Department of Biology, University of Puerto Rico in Ponce, Ponce, Puerto Rico
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland, United States
- Biophysics Program, University of Maryland, College Park, Maryland, United States
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, Maryland, United States
| |
Collapse
|
2
|
Khan A, Ni W, Baltazar T, Lopez-Giraldez F, Pober JS, Pierce RW. ArhGEF12 activates Rap1A and not RhoA in human dermal microvascular endothelial cells to reduce tumor necrosis factor-induced leak. FASEB J 2022; 36:e22254. [PMID: 35294066 PMCID: PMC9103844 DOI: 10.1096/fj.202101873rr] [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: 12/06/2021] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 12/31/2022]
Abstract
Overwhelming inflammation in the setting of acute critical illness induces capillary leak resulting in hypovolemia, edema, tissue dysoxia, organ failure and even death. The tight junction (TJ)-dependent capillary barrier is regulated by small GTPases, but the specific regulatory molecules most active in this vascular segment under such circumstances are not well described. We set out to identify GTPase regulatory molecules specific to endothelial cells (EC) that form TJs. Transcriptional profiling of confluent monolayers of TJ-forming human dermal microvascular ECs (HDMECs) and adherens junction only forming-human umbilical vein EC (HUVECs) demonstrate ARHGEF12 is basally expressed at higher levels and is only downregulated in HDMECs by junction-disrupting tumor necrosis factor (TNF). HDMECs depleted of ArhGEF12 by siRNA demonstrate a significantly exacerbated TNF-induced decrease in trans-endothelial electrical resistance and disruption of TJ continuous staining. ArhGEF12 is established as a RhoA-GEF in HUVECs and its knock down would be expected to reduce RhoA activity and barrier disruption. Pulldown of active GEFs from HDMECs depleted of ArhGEF12 and treated with TNF show decreased GTP-bound Rap1A after four hours but increased GTP-bound RhoA after 12 h. In cell-free assays, ArhGEF12 immunoprecipitated from HDMECs is able to activate both Rap1A and RhoA, but not act on Rap2A-C, RhoB-C, or even Rap1B which shares 95% sequence identity with Rap1A. We conclude that in TJ-forming HDMECs, ArhGEF12 selectively activates Rap1A to limit capillary barrier disruption in a mechanism independent of cAMP-mediated Epac1 activation.
Collapse
Affiliation(s)
- Alamzeb Khan
- Department of Pediatrics, Yale School of Medicine, Yale University
| | - Weiming Ni
- Department of Pediatrics, Yale School of Medicine, Yale University
| | - Tania Baltazar
- Department of Immunobiology, Yale School of Medicine, Yale University
| | | | - Jordan S. Pober
- Department of Immunobiology, Yale School of Medicine, Yale University
| | | |
Collapse
|
3
|
Smyth T, Georas SN. Effects of ozone and particulate matter on airway epithelial barrier structure and function: a review of in vitro and in vivo studies. Inhal Toxicol 2021; 33:177-192. [PMID: 34346824 DOI: 10.1080/08958378.2021.1956021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The airway epithelium represents a crucial line of defense against the spread of inhaled pathogens. As the epithelium is the first part of the body to be exposed to the inhaled environment, it must act as both a barrier to and sentinel against any inhaled agents. Despite its vital role in limiting the spread of inhaled pathogens, the airway epithelium is also regularly exposed to air pollutants which disrupt its normal function. Here we review the current understanding of the structure and composition of the airway epithelial barrier, as well as the impact of inhaled pollutants, including the reactive gas ozone and particulate matter, on epithelial function. We discuss the current in vitro, rodent model, and human exposure findings surrounding the impact of various inhaled pollutants on epithelial barrier function, mucus production, and mucociliary clearance. Detailed information on how inhaled pollutants impact epithelial structure and function will further our understanding of the adverse health effects of air pollution exposure.
Collapse
Affiliation(s)
- Timothy Smyth
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Steve N Georas
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
4
|
Augustine R, Zahid AA, Mraiche F, Alam K, Al Moustafa AE, Hasan A. Gelatin-methacryloyl hydrogel based in vitro blood-brain barrier model for studying breast cancer-associated brain metastasis. Pharm Dev Technol 2021; 26:490-500. [PMID: 33416013 DOI: 10.1080/10837450.2021.1872624] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breast cancer is one of the leading causes of brain metastasis. Metastasis to the brain occurs if cancer cells manage to traverse the 'blood-brain barrier' (BBB), which is a barrier with a very tight junction (TJ) of endothelial cells between blood circulation and brain tissue. It is highly important to develop novel in vitro BBB models to investigate breast cancer metastasis to the brain to facilitate the screening of chemotherapeutic agents against it. We herein report the development of gelatin methacryloyl (GelMA) modified transwell insert based BBB model composed of endothelial and astrocyte cell layers for testing the efficacy of anti-metastatic agents against breast cancer metastasis to the brain. We characterized the developed model for the morphology and in vitro breast cancer cell migration. Furthermore, we investigated the effect of cisplatin, a widely used chemotherapeutic agent, on the migration of metastatic breast cancer cells using the model. Our results showed that breast cancer cells migrate across the developed BBB model. Cisplatin treatment inhibited the migration of cancer cells across the model. Findings of this study suggest that our BBB model can be used as a suitable tool to investigate breast cancer-associated brain metastasis and to identify suitable therapeutic agents against this.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar.,Biomedical Research Center (BRC), Qatar University, Doha, Qatar
| | - Alap Ali Zahid
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar.,Biomedical Research Center (BRC), Qatar University, Doha, Qatar
| | - Fatima Mraiche
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Khurshid Alam
- Department of Mechanical and Industrial Engineering, College of Engineering, Sultan Qaboos University, Sultanate of Oman
| | - Ala-Eddin Al Moustafa
- Biomedical Research Center (BRC), Qatar University, Doha, Qatar.,College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar.,Biomedical Research Center (BRC), Qatar University, Doha, Qatar
| |
Collapse
|
5
|
Abstract
Neutrophils have always been considered as uncomplicated front-line troopers of the innate immune system equipped with limited proinflammatory duties. Yet recently, the role of the neutrophil has been undergoing a rejuvenation of sorts. Neutrophils are now considered complex cells capable of a significant array of specialized functions, and as an effector of the innate immune response, they are able to regulate many processes such as acute injury and repair, cancer, autoimmunity, and chronic inflammatory processes. Furthermore, evidence exists to indicate that neutrophils also contribute to adaptive immunity by aiding the development of specific adaptive immune responses or guiding the subsequent adaptive immune response. With this revived interest in neutrophils and their many novel functions, it is prudent to review what is currently known about neutrophils and, even more importantly, understand what information is lacking. We discuss the essential features of the neutrophil, from its origins, lifespan, subsets, margination and sequestration of the neutrophil to the death of the neutrophil. We highlight neutrophil recruitment to both infected and injured tissues and outline differences in recruitment of neutrophils between different tissues. Finally, we examine how neutrophils use different mechanisms to either bolster protective immune responses or negatively cause pathological outcomes at different locations.
Collapse
Affiliation(s)
- Pei Xiong Liew
- Snyder Institute of Chronic Diseases, University of Calgary, Calgary, Alberta, Canada; and Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Paul Kubes
- Snyder Institute of Chronic Diseases, University of Calgary, Calgary, Alberta, Canada; and Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
6
|
Shi J, Barakat M, Chen D, Chen L. Bicellular Tight Junctions and Wound Healing. Int J Mol Sci 2018; 19:ijms19123862. [PMID: 30518037 PMCID: PMC6321209 DOI: 10.3390/ijms19123862] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 12/15/2022] Open
Abstract
Bicellular tight junctions (TJs) are intercellular junctions comprised of a variety of transmembrane proteins including occludin, claudins, and junctional adhesion molecules (JAMs) as well as intracellular scaffold proteins such as zonula occludens (ZOs). TJs are functional, intercellular structures that form a barrier between adjacent cells, which constantly seals and unseals to control the paracellular passage of molecules. They are primarily present in the epithelial and endothelial cells of all tissues and organs. In addition to their well-recognized roles in maintaining cell polarity and barrier functions, TJs are important regulators of signal transduction, which modulates cell proliferation, migration, and differentiation, as well as some components of the immune response and homeostasis. A vast breadth of research data is available on TJs, but little has been done to decipher their specific roles in wound healing, despite their primary distribution in epithelial and endothelial cells, which are essential contributors to the wound healing process. Some data exists to indicate that a better understanding of the functions and significance of TJs in healing wounds may prove crucial for future improvements in wound healing research and therapy. Specifically, recent studies demonstrate that occludin and claudin-1, which are two TJ component proteins, are present in migrating epithelial cells at the wound edge but are absent in chronic wounds. This indicates that functional TJs may be critical for effective wound healing. A tremendous amount of work is needed to investigate their roles in barrier function, re-epithelialization, angiogenesis, scar formation, and in the interactions between epithelial cells, endothelial cells, and immune cells both in the acute wound healing process and in non-healing wounds. A more thorough understanding of TJs in wound healing may shed new light on potential research targets and reveal novel strategies to enhance tissue regeneration and improve wound repair.
Collapse
Affiliation(s)
- Junhe Shi
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, 801 S. Paulina Street, Chicago, IL 60612, USA.
| | - May Barakat
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, 801 S. Paulina Street, Chicago, IL 60612, USA.
| | - Dandan Chen
- Colgate-Palmolive Company, Piscataway, NJ 08855, USA.
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, 801 S. Paulina Street, Chicago, IL 60612, USA.
| |
Collapse
|
7
|
Diedrichs-Möhring M, Kaufmann U, Wildner G. The immunopathogenesis of chronic and relapsing autoimmune uveitis – Lessons from experimental rat models. Prog Retin Eye Res 2018; 65:107-126. [DOI: 10.1016/j.preteyeres.2018.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/19/2018] [Accepted: 02/22/2018] [Indexed: 12/12/2022]
|
8
|
Alvarenga DM, Mattos MS, Araújo AM, Antunes MM, Menezes GB. Neutrophil biology within hepatic environment. Cell Tissue Res 2017; 371:589-598. [PMID: 29127519 DOI: 10.1007/s00441-017-2722-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/26/2017] [Indexed: 12/29/2022]
Abstract
Neutrophils are the most abundant leukocyte in the human circulation. These short-lived cells are constantly produced from hematopoietic stem cells (HSC) within the bone marrow from which they daily reach the blood and perform major roles in innate immunity. Neutrophils are the first cells to reach inflamed tissues and are armed with a plethora of enzymes that help both with their trafficking within tissues and the killing of pathogens. Damaged or infected organs are rapidly invaded by neutrophils. Their erroneous activation within parenchyma or the vasculature is involved in the pathogenesis of several inflammatory diseases including arthritis, colitis, sepsis, acute lung injury and liver failure. Despite the proposal of a canonical pathway that governs neutrophil migration into tissues, the liver has been extensively described as a unique environment for leukocyte recruitment. Since the control of inflammatory responses is considered one of the most promising avenues for novel therapeutics, the expansion of our understanding of the mechanisms behind neutrophil accumulation within injured liver might add to the development of specific and more efficacious treatments. In this review, we discuss the basic concepts of neutrophil ontogeny and biology, with a focus on the particularities and the molecular steps involved in neutrophil recruitment to the liver.
Collapse
Affiliation(s)
- Débora Moreira Alvarenga
- Center for Gastrointestinal Biology, Departamento de Morfologia, Sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. PresidenteAntônio Carlos, 6627 - Pampulha, Belo Horizonte, Minas Gerais, Brasil
| | - Matheus Silvério Mattos
- Center for Gastrointestinal Biology, Departamento de Morfologia, Sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. PresidenteAntônio Carlos, 6627 - Pampulha, Belo Horizonte, Minas Gerais, Brasil
| | - Alan Moreira Araújo
- Center for Gastrointestinal Biology, Departamento de Morfologia, Sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. PresidenteAntônio Carlos, 6627 - Pampulha, Belo Horizonte, Minas Gerais, Brasil
| | - Maísa Mota Antunes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. PresidenteAntônio Carlos, 6627 - Pampulha, Belo Horizonte, Minas Gerais, Brasil.
| | - Gustavo Batista Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. PresidenteAntônio Carlos, 6627 - Pampulha, Belo Horizonte, Minas Gerais, Brasil.
| |
Collapse
|
9
|
Abstract
Myeloid cell recruitment to sites of infection and injury started out as a simple model that has been referred to as the universal concept of leukocyte recruitment. However, as we gain more insight into the different mechanisms, it is becoming clear that each organ and perhaps even each cell has its own unique mechanism of recruitment. Moreover, as the ability to visualize specific cell types in specific organs becomes more accessible, it is also becoming clear that there are resident populations of leukocytes, some within the tissues and others attached to the vasculature of tissues, the latter poised to affect the local environment. In this review, we will first highlight the imaging approaches that have allowed us to gain spectacular insight into locale and function of specific cell types, and then we will discuss what we have learned from this approach as far as myeloid cells are concerned. We will also highlight some of the gaps in our knowledge, which exist almost certainly because of the challenges of being able to visualize certain compartments of the body.
Collapse
|
10
|
Leukocyte Kinetics and Migration in the Lungs. Respir Med 2017. [DOI: 10.1007/978-3-319-41912-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
11
|
Teijeira A, Halin C. Editorial: Breaching their way through: Neutrophils destroy intercellular junctions to transmigrate rapidly across lymphatic endothelium. J Leukoc Biol 2016; 98:880-2. [PMID: 26628638 DOI: 10.1189/jlb.3ce0615-273] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Alvaro Teijeira
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Fromen CA, Fish MB, Zimmerman A, Adili R, Holinstat M, Eniola-Adefeso O. Evaluation of Receptor-Ligand Mechanisms of Dual-Targeted Particles to an Inflamed Endothelium. Bioeng Transl Med 2016; 1:103-115. [PMID: 28066821 PMCID: PMC5217161 DOI: 10.1002/btm2.10008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 04/25/2016] [Accepted: 04/27/2016] [Indexed: 12/11/2022] Open
Abstract
Vascular-targeted carriers (VTCs) are designed as leukocyte mimics, decorated with ligands that target leukocyte adhesion molecules (LAMs) and facilitate adhesion to diseased endothelium. VTCs require different design considerations than other targeted particle therapies; adhesion of VTCs in regions with dynamic blood flow requires multiple ligand-receptor (LR) pairs that provide particle adhesion and disease specificity. Despite the ultimate goal of leukocyte mimicry, the specificity of multiple LAM-targeted VTCs remains poorly understood, especially in physiological environments. Here, we investigate particle binding to an inflamed mesentery via intravital microscopy using a series of particles with well-controlled ligand properties. We find that the total number of sites of a single ligand can drive particle adhesion to the endothelium, however, combining ligands that target multiple LR pairs provides a more effective approach. Combining sites of sialyl Lewis A (sLeA) and anti-intercellular adhesion molecule-1 (aICAM), two adhesive molecules, resulted in ~3-7-fold increase of adherent particles at the endothelium over single-ligand particles. At a constant total ligand density, a particle with a ratio of 75% sLeA: 25% aICAM resulted in more than 3-fold increase over all over other ligand ratios tested in our in vivo model. Combined with in vitro and in silico data, we find the best dual-ligand design of a particle is heavily dependent on the surface expression of the endothelial cells, producing better adhesion with more particle ligand for the lesser-expressed receptor. These results establish the importance of considering LR-kinetics in intelligent VTC ligand design for future therapeutics.
Collapse
Affiliation(s)
- Catherine A Fromen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Margaret B Fish
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Anthony Zimmerman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Reheman Adili
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109; Division of Cardiovascular Medicine, Samuel and Jean Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109
| | | |
Collapse
|
13
|
Hou J, Baker LA, Zhou L, Klein RS. Viral interactions with the blood-brain barrier: old dog, new tricks. Tissue Barriers 2016; 4:e1142492. [PMID: 27141421 DOI: 10.1080/21688370.2016.1142492] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/05/2016] [Accepted: 01/07/2016] [Indexed: 02/07/2023] Open
Abstract
Brain endothelial cells form a unique cellular structure known as the tight junction to regulate the exchanges between the blood and the parenchyma by limiting the paracellular diffusion of blood-borne substance. Together with the restricted pathway of transcytosis, the tight junction in the brain endothelial cells provides the central nervous system (CNS) with effective protection against both the foreign pathogens and the host immune cells, which is also termed the "blood-brain barrier." The blood-brain barrier is particularly important for defending against neurotropic viral infections that have become a major source of diseases worldwide. Many neurotropic viruses are able to cross the BBB and infect the CNS through very poorly understood processes. This review focuses upon the structural and functional changes of the brain endothelial tight junction in response to viral infections in the CNS and how the tight junction changes may be studied with advanced imaging and recording approaches to reveal novel processes used by the viruses to cross the barrier system. Additional emphasis is placed upon new countermeasures that can act directly upon the tight junction to improve the pathogen clearance and minimize the inflammatory damage.
Collapse
Affiliation(s)
- Jianghui Hou
- Internal Medicine, Washington University in St Louis; St. Louis, MO USA; Center for Investigation of Membrane Excitability Diseases, Washington University in St Louis; St. Louis, MO USA
| | - Lane A Baker
- Department of Chemistry; Indiana University ; Bloomington, IN USA
| | - Lushan Zhou
- Department of Chemistry; Indiana University ; Bloomington, IN USA
| | - Robyn S Klein
- Internal Medicine, Washington University in St Louis; St. Louis, MO USA; Anatomy and Neurobiology, Washington University in St Louis; St. Louis, MO USA; Pathology and Immunology, Washington University in St Louis; St. Louis, MO USA
| |
Collapse
|
14
|
Schossleitner K, Rauscher S, Gröger M, Friedl HP, Finsterwalder R, Habertheuer A, Sibilia M, Brostjan C, Födinger D, Citi S, Petzelbauer P. Evidence That Cingulin Regulates Endothelial Barrier Function In Vitro and In Vivo. Arterioscler Thromb Vasc Biol 2016; 36:647-54. [PMID: 26821949 DOI: 10.1161/atvbaha.115.307032] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 01/14/2016] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Cingulin is a cytoplasmic component of tight junctions. Although modulation of cingulin levels in cultured epithelial model systems has no significant effect on barrier function, evidence from cingulin knockout mice suggests that cingulin may be involved in the regulation of the behavior of epithelial or endothelial cells. Here, we investigate the role of cingulin in the barrier function of endothelial cells. APPROACH AND RESULTS We show that cingulin is expressed in human endothelial cells of the skin, brain, and lung in vivo and in vitro. Endothelial cingulin colocalizes and coimmunoprecipitates with the tight junction proteins zonula occludens-1 and guanine nucleotide exchange factor-H1. Cingulin overexpression in human umbilical vein endothelial cell induces tight junction formation, increases transendothelial electric resistance, and strengthens barrier function for low and high molecular weight tracers. In contrast, cultured endothelial cells lacking cingulin are more permeable for low molecular weight tracers. In cingulin knockout mice, neurons of the area postrema and Purkinje cells show an increased uptake of small molecular weight tracers indicating decreased barrier function at these sites. CONCLUSIONS We demonstrate that cingulin participates in the modulation of endothelial barrier function both in human cultured cells in vitro and in mouse brains in vivo. Understanding the role of cingulin in maintaining tight barriers in endothelia may allow developing new strategies for the treatment of vascular leak syndromes.
Collapse
Affiliation(s)
- Klaudia Schossleitner
- From the Skin and Endothelium Research Division (SERD), Department of Dermatology (K.S., S.R., M.G., H.P.F., R.F., P.P.), Core Facility Imaging (S.R., M.G.), Department of Cardiac Surgery (A.H.), Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center (M.S.), Department of Surgery (C.B.), and Department of Dermatology (D.F.), Medical University of Vienna, Vienna, Austria; and Department of Cell Biology and Institute of Genetics and Genomics in Geneva, University of Geneva, Switzerland (S.C.)
| | - Sabine Rauscher
- From the Skin and Endothelium Research Division (SERD), Department of Dermatology (K.S., S.R., M.G., H.P.F., R.F., P.P.), Core Facility Imaging (S.R., M.G.), Department of Cardiac Surgery (A.H.), Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center (M.S.), Department of Surgery (C.B.), and Department of Dermatology (D.F.), Medical University of Vienna, Vienna, Austria; and Department of Cell Biology and Institute of Genetics and Genomics in Geneva, University of Geneva, Switzerland (S.C.)
| | - Marion Gröger
- From the Skin and Endothelium Research Division (SERD), Department of Dermatology (K.S., S.R., M.G., H.P.F., R.F., P.P.), Core Facility Imaging (S.R., M.G.), Department of Cardiac Surgery (A.H.), Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center (M.S.), Department of Surgery (C.B.), and Department of Dermatology (D.F.), Medical University of Vienna, Vienna, Austria; and Department of Cell Biology and Institute of Genetics and Genomics in Geneva, University of Geneva, Switzerland (S.C.)
| | - Heinz Peter Friedl
- From the Skin and Endothelium Research Division (SERD), Department of Dermatology (K.S., S.R., M.G., H.P.F., R.F., P.P.), Core Facility Imaging (S.R., M.G.), Department of Cardiac Surgery (A.H.), Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center (M.S.), Department of Surgery (C.B.), and Department of Dermatology (D.F.), Medical University of Vienna, Vienna, Austria; and Department of Cell Biology and Institute of Genetics and Genomics in Geneva, University of Geneva, Switzerland (S.C.)
| | - Richard Finsterwalder
- From the Skin and Endothelium Research Division (SERD), Department of Dermatology (K.S., S.R., M.G., H.P.F., R.F., P.P.), Core Facility Imaging (S.R., M.G.), Department of Cardiac Surgery (A.H.), Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center (M.S.), Department of Surgery (C.B.), and Department of Dermatology (D.F.), Medical University of Vienna, Vienna, Austria; and Department of Cell Biology and Institute of Genetics and Genomics in Geneva, University of Geneva, Switzerland (S.C.)
| | - Andreas Habertheuer
- From the Skin and Endothelium Research Division (SERD), Department of Dermatology (K.S., S.R., M.G., H.P.F., R.F., P.P.), Core Facility Imaging (S.R., M.G.), Department of Cardiac Surgery (A.H.), Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center (M.S.), Department of Surgery (C.B.), and Department of Dermatology (D.F.), Medical University of Vienna, Vienna, Austria; and Department of Cell Biology and Institute of Genetics and Genomics in Geneva, University of Geneva, Switzerland (S.C.)
| | - Maria Sibilia
- From the Skin and Endothelium Research Division (SERD), Department of Dermatology (K.S., S.R., M.G., H.P.F., R.F., P.P.), Core Facility Imaging (S.R., M.G.), Department of Cardiac Surgery (A.H.), Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center (M.S.), Department of Surgery (C.B.), and Department of Dermatology (D.F.), Medical University of Vienna, Vienna, Austria; and Department of Cell Biology and Institute of Genetics and Genomics in Geneva, University of Geneva, Switzerland (S.C.)
| | - Christine Brostjan
- From the Skin and Endothelium Research Division (SERD), Department of Dermatology (K.S., S.R., M.G., H.P.F., R.F., P.P.), Core Facility Imaging (S.R., M.G.), Department of Cardiac Surgery (A.H.), Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center (M.S.), Department of Surgery (C.B.), and Department of Dermatology (D.F.), Medical University of Vienna, Vienna, Austria; and Department of Cell Biology and Institute of Genetics and Genomics in Geneva, University of Geneva, Switzerland (S.C.)
| | - Dagmar Födinger
- From the Skin and Endothelium Research Division (SERD), Department of Dermatology (K.S., S.R., M.G., H.P.F., R.F., P.P.), Core Facility Imaging (S.R., M.G.), Department of Cardiac Surgery (A.H.), Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center (M.S.), Department of Surgery (C.B.), and Department of Dermatology (D.F.), Medical University of Vienna, Vienna, Austria; and Department of Cell Biology and Institute of Genetics and Genomics in Geneva, University of Geneva, Switzerland (S.C.)
| | - Sandra Citi
- From the Skin and Endothelium Research Division (SERD), Department of Dermatology (K.S., S.R., M.G., H.P.F., R.F., P.P.), Core Facility Imaging (S.R., M.G.), Department of Cardiac Surgery (A.H.), Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center (M.S.), Department of Surgery (C.B.), and Department of Dermatology (D.F.), Medical University of Vienna, Vienna, Austria; and Department of Cell Biology and Institute of Genetics and Genomics in Geneva, University of Geneva, Switzerland (S.C.)
| | - Peter Petzelbauer
- From the Skin and Endothelium Research Division (SERD), Department of Dermatology (K.S., S.R., M.G., H.P.F., R.F., P.P.), Core Facility Imaging (S.R., M.G.), Department of Cardiac Surgery (A.H.), Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center (M.S.), Department of Surgery (C.B.), and Department of Dermatology (D.F.), Medical University of Vienna, Vienna, Austria; and Department of Cell Biology and Institute of Genetics and Genomics in Geneva, University of Geneva, Switzerland (S.C.).
| |
Collapse
|
15
|
Wu X, Newbold MA, Haynes CL. Recapitulation of in vivo-like neutrophil transendothelial migration using a microfluidic platform. Analyst 2016; 140:5055-64. [PMID: 26087389 DOI: 10.1039/c5an00967g] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Neutrophil transendothelial migration (TEM) is an essential physiological process that regulates the recruitment of neutrophils in response to inflammatory signals. Herein, a versatile hydrogel scaffold is embedded in a microfluidic platform that supports an endothelial cell layer cultured in the vertical direction and highly stable chemical gradients; this construct is employed to mimic the in vivo neutrophil TEM process. We found that the number of neutrophils migrating across the endothelial cell layer is dependent on the presented chemoattractant concentration and the spatial profile of the chemical gradient. Endothelial cells play a critical role in neutrophil TEM by promoting neutrophil morphological changes as well as expressing surface receptor molecules that are indispensable for inducing neutrophil attachment and migration. Furthermore, the microfluidic device also supports competing chemoattractant gradients to facilitate neutrophil TEM studies in complex microenvironments that more accurately model the in vivo system than simplified microenvironments without the complexity of chemical gradients. This work demonstrates that combinations of any two different chemoattractants induce more significant neutrophil migration than a single chemoattractant in the same total amount, indicating synergistic effects between distinct chemoattractants. The in vitro reconstitution of neutrophil TEM successfully translates planar neutrophil movement into in vivo-like neutrophil recruitment and accelerates understanding of cellular interactions between neutrophils and endothelial cells within the complicated physiological milieu.
Collapse
Affiliation(s)
- Xiaojie Wu
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, USA.
| | | | | |
Collapse
|
16
|
Namdee K, Sobczynski DJ, Onyskiw PJ, Eniola-Adefeso O. Differential Impact of Plasma Proteins on the Adhesion Efficiency of Vascular-Targeted Carriers (VTCs) in Blood of Common Laboratory Animals. Bioconjug Chem 2015; 26:2419-28. [PMID: 26505780 PMCID: PMC4866610 DOI: 10.1021/acs.bioconjchem.5b00474] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Vascular-targeted carrier (VTC) interaction with human plasma is known to reduce targeted adhesion efficiency in vitro. However, the role of plasma proteins on the adhesion efficiency of VTCs in laboratory animals remains unknown. Here, in vitro blood flow assays are used to explore the effects of plasma from mouse, rabbit, and porcine on VTC adhesion. Porcine blood exhibited a strong negative plasma effect on VTC adhesion while no significant plasma effect was found with rabbit and mouse blood. A brush density poly(ethylene glycol) (PEG) on VTCs was effective at improving adhesion of microsized, but not nanosized, VTCs in porcine blood. Overall, the results suggest that porcine models, as opposed to mouse, can serve as better models in preclinical research for predicting the in vivo functionality of VTCs for use in humans. These considerations hold great importance for the design of various pharmaceutical products and development of reliable drug delivery systems.
Collapse
Affiliation(s)
| | | | - Peter J. Onyskiw
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109
| |
Collapse
|
17
|
Tajes M, Ramos-Fernández E, Weng-Jiang X, Bosch-Morató M, Guivernau B, Eraso-Pichot A, Salvador B, Fernàndez-Busquets X, Roquer J, Muñoz FJ. The blood-brain barrier: structure, function and therapeutic approaches to cross it. Mol Membr Biol 2014; 31:152-67. [PMID: 25046533 DOI: 10.3109/09687688.2014.937468] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The blood-brain barrier (BBB) is constituted by a specialized vascular endothelium that interacts directly with astrocytes, neurons and pericytes. It protects the brain from the molecules of the systemic circulation but it has to be overcome for the proper treatment of brain cancer, psychiatric disorders or neurodegenerative diseases, which are dramatically increasing as the population ages. In the present work we have revised the current knowledge on the cellular structure of the BBB and the different procedures utilized currently and those proposed to cross it. Chemical modifications of the drugs, such as increasing their lipophilicity, turn them more prone to be internalized in the brain. Other mechanisms are the use of molecular tools to bind the drugs such as small immunoglobulins, liposomes or nanoparticles that will act as Trojan Horses favoring the drug delivery in brain. This fusion of the classical pharmacology with nanotechnology has opened a wide field to many different approaches with promising results to hypothesize that BBB will not be a major problem for the new generation of neuroactive drugs. The present review provides an overview of all state-of-the-art of the BBB structure and function, as well as of the classic strategies and these appeared in recent years to deliver drugs into the brain for the treatment of Central Nervous System (CNS) diseases.
Collapse
Affiliation(s)
- Marta Tajes
- Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF) , Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Affiliation(s)
- Jing Wang
- Department of Physiology and Pharmacology; University of Calgary; Calgary Alberta Canada
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases; University of Calgary; Calgary Alberta Canada
- Department of Immunochemistry; Research Institute for Microbial Diseases; Osaka University; Osaka Japan
| | - Hisashi Arase
- Department of Immunochemistry; Research Institute for Microbial Diseases; Osaka University; Osaka Japan
- Laboratory of Immunochemistry; World Premier International Immunology Frontier Research Center; Osaka University; Osaka Japan
- Core Research for Evolutional Science and Technology; Japan Science and Technology Agency; Saitama Japan
| |
Collapse
|
19
|
Tossetta G, Paolinelli F, Avellini C, Salvolini E, Ciarmela P, Lorenzi T, Emanuelli M, Toti P, Giuliante R, Gesuita R, Crescimanno C, Voltolini C, Di Primio R, Petraglia F, Castellucci M, Marzioni D. IL-1β and TGF-β weaken the placental barrier through destruction of tight junctions: an in vivo and in vitro study. Placenta 2014; 35:509-16. [PMID: 24768095 DOI: 10.1016/j.placenta.2014.03.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 03/19/2014] [Accepted: 03/24/2014] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Chorioamnionitis is a gestational pathological condition characterized by acute inflammation of the amniochorionic membranes and placentas leading to high concentrations of IL-1β, Il-6, Il-8 and TGF-β in the amniotic fluid. In normal conditions, the permeability of foeto-maternal barrier is due to the assembly and maintenance of different cellular junctional domains. METHODS In the present study, first we aimed to evaluate the protein expression (by immunohistochemistry and western blotting) and mRNA (by real time PCR) levels of the molecular components of tight junctions (Zonula occludens-1 and occludin), and of adherent junctions (VE-cadherin and β-catenin) in placentas from chorioamnionitis compared to that in normal pregnancies. RESULTS Western blotting results showed a significant down-regulation of occludin in placentas affected with chorioamnionitis. No differences were detected for the other proteins analysed. We evaluated whether occludin expression was regulated by IL-1β, IL-6, IL-8 and TGF-β by means of in vitro studies using HUVEC cultures and demonstrated a key role of IL-1β and TGF-β in the disappearance of occludin at cellular border. CONCLUSIONS We conclude by suggesting a pivotal role of these two cytokines in facilitating intra-placental infection via para-cellular way due to the disassembly of tight junctions at trophoblastic and endothelial cells in placental tissues.
Collapse
Affiliation(s)
- G Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60020 Ancona, Italy.
| | - F Paolinelli
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60020 Ancona, Italy.
| | - C Avellini
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60020 Ancona, Italy.
| | - E Salvolini
- Department of Molecular and Clinical Sciences-Histology, Università Politecnica delle Marche, 60020 Ancona, Italy.
| | - P Ciarmela
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60020 Ancona, Italy.
| | - T Lorenzi
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60020 Ancona, Italy.
| | - M Emanuelli
- Department of Clinical Sciences, Section of Biochemistry, Università Politecnica delle Marche, 60131 Ancona, Italy.
| | - P Toti
- Department of Medical Biotechnologies, Pathology Unit, University of Siena, Siena, Italy.
| | - R Giuliante
- Department of Clinical Sciences, Section of Biochemistry, Università Politecnica delle Marche, 60131 Ancona, Italy.
| | - R Gesuita
- Department of Biomedical Sciences and Public Health, Epidemiological and Bio-statistic Centre, Università Politecnica delle Marche, 60020 Ancona, Italy.
| | - C Crescimanno
- Faculty of Engineering, Architecture and Physical Education, Università Kore, 94100 Enna, Italy.
| | - C Voltolini
- Department of Molecular and Developmental Medicine-Obstetrics and Gynecology, University of Siena, 53100 Siena, Italy.
| | - R Di Primio
- Department of Molecular and Clinical Sciences-Histology, Università Politecnica delle Marche, 60020 Ancona, Italy.
| | - F Petraglia
- Department of Molecular and Developmental Medicine-Obstetrics and Gynecology, University of Siena, 53100 Siena, Italy.
| | - M Castellucci
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60020 Ancona, Italy.
| | - D Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60020 Ancona, Italy.
| |
Collapse
|
20
|
Liggett JL, Choi CK, Donnell RL, Kihm KD, Kim JS, Min KW, Noegel AA, Baek SJ. Nonsteroidal anti-inflammatory drug sulindac sulfide suppresses structural protein Nesprin-2 expression in colorectal cancer cells. Biochim Biophys Acta Gen Subj 2013; 1840:322-31. [PMID: 24080406 DOI: 10.1016/j.bbagen.2013.09.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 08/22/2013] [Accepted: 09/20/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND Nonsteroidal anti-inflammatory drugs (NSAIDs) are well known for treating inflammatory disease and have been reported to have anti-tumorigenic effects. Their mechanisms are not fully understood, but both cyclooxygenase (COX) dependent and independent pathways are involved. Our goal was to shed further light on COX-independent activity. METHODS Human colorectal cancer cells were observed under differential interference contrast microscopy (DICM), fluorescent microscopy, and micro-impedance measurement. Microarray analysis was performed using HCT-116 cells treated with sulindac sulfide (SS). PCR and Western blots were performed to confirm the microarray data and immunohistochemistry was performed to screen for Nesprin-2 expression. Micro-impedance was repeating including Nesprin-2 knock-down by siRNA. RESULTS HCT-116 cells treated with SS showed dramatic morphological changes under DICM and fluorescent microscopy, as well as weakened cellular adhesion as measured by micro-impedance. Nesprin-2 was selected from two independent microarrays, based on its novelty in relation to cancer and its role in cell organization. SS diminished Nesprin-2 mRNA expression as assessed by reverse transcriptase and real time PCR. Various other NSAIDs were also tested and demonstrated that inhibition of Nesprin-2 mRNA was not unique to SS. Additionally, immunohistochemistry showed higher levels of Nesprin-2 in many tumors in comparison with normal tissues. Further micro-impedance experiments on cells with reduced Nesprin-2 expression showed a proportional loss of cellular adhesion. CONCLUSIONS Nesprin-2 is down-regulated by NSAIDs and highly expressed in many cancers. GENERAL SIGNIFICANCE Our data suggest that Nesprin-2 may be a potential novel oncogene in human cancer cells and NSAIDs could decrease its expression.
Collapse
Affiliation(s)
- Jason L Liggett
- Department of Biomedical and Diagnostic Sciences, University of Tennessee, Knoxville, TN 37996, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Onyskiw PJ, Eniola-Adefeso O. Effect of PEGylation on ligand-based targeting of drug carriers to the vascular wall in blood flow. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2013; 29:11127-34. [PMID: 23919766 DOI: 10.1021/la402182j] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
The blood vessel wall plays a prominent role in the development of many life-threatening diseases and as such is an attractive target for treatment. To target diseased tissue, particulate drug carriers often have their surfaces modified with antibodies or epitopes specific to vascular wall-expressed molecules, along with poly(ethylene glycol) (PEG) to improve carrier blood circulation time. However, little is known about the effect of poly(ethylene glycol) on carrier adhesion dynamics-specifically in blood flow. Here we examine the influence of different molecular weight PEG spacers on particle adhesion in blood flow. Anti-ICAM-1 or Sialyl Lewis(a) were grafted onto polystyrene 2 μm and 500 nm spheres via PEG spacers and perfused in blood over activated endothelial cells at physiological shear conditions. PEG spacers were shown to improve, reduce, or have no effect on the binding density of targeted-carriers depending on the PEG surface conformation, shear rate, and targeting moiety.
Collapse
Affiliation(s)
- Peter J Onyskiw
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | |
Collapse
|
22
|
Abstract
Neutrophils have traditionally been thought of as simple foot soldiers of the innate immune system with a restricted set of pro-inflammatory functions. More recently, it has become apparent that neutrophils are, in fact, complex cells capable of a vast array of specialized functions. Although neutrophils are undoubtedly major effectors of acute inflammation, several lines of evidence indicate that they also contribute to chronic inflammatory conditions and adaptive immune responses. Here, we discuss the key features of the life of a neutrophil, from its release from bone marrow to its death. We discuss the possible existence of different neutrophil subsets and their putative anti-inflammatory roles. We focus on how neutrophils are recruited to infected or injured tissues and describe differences in neutrophil recruitment between different tissues. Finally, we explain the mechanisms that are used by neutrophils to promote protective or pathological immune responses at different sites.
Collapse
|
23
|
Transendothelial migration enables subsequent transmigration of neutrophils through underlying pericytes. PLoS One 2013; 8:e60025. [PMID: 23555870 PMCID: PMC3608600 DOI: 10.1371/journal.pone.0060025] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 02/21/2013] [Indexed: 01/13/2023] Open
Abstract
During acute inflammation, neutrophil recruitment into extravascular tissue requires neutrophil tethering and rolling on cytokine-activated endothelial cells (ECs), tight adhesion, crawling towards EC junctions and transendothelial migration (TEM). Following TEM, neutrophils must still traverse the subendothelial basement membrane and network of pericytes (PCs). Until recently, the contribution of the PC layer to neutrophil recruitment was largely ignored. Here we analyze human neutrophil interactions with interleukin (IL)-1β-activated human EC monolayers, PC monolayers and EC/PC bilayers in vitro. Compared to EC, PC support much lower levels of neutrophil binding (54.6% vs. 7.1%, respectively) and transmigration (63.7 vs. 8.8%, respectively) despite comparable levels of IL-8 (CXCL8) synthesis and display. Remarkably, EC/PC bilayers support intermediate levels of transmigration (37.7%). Neutrophil adhesion to both cell types is Mac-1-dependent and while ICAM-1 transduction of PCs increases neutrophil adhesion to (41.4%), it does not increase transmigration through PC monolayers. TEM, which increases neutrophil Mac-1 surface expression, concomitantly increases the ability of neutrophils to traverse PCs (19.2%). These data indicate that contributions from both PCs and ECs must be considered in evaluation of microvasculature function in acute inflammation.
Collapse
|
24
|
Transmigration of polymorphnuclear neutrophils and monocytes through the human blood-cerebrospinal fluid barrier after bacterial infection in vitro. J Neuroinflammation 2013; 10:31. [PMID: 23448224 PMCID: PMC3663685 DOI: 10.1186/1742-2094-10-31] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 02/18/2013] [Indexed: 01/13/2023] Open
Abstract
Background Bacterial invasion through the blood-cerebrospinal fluid barrier (BCSFB) during bacterial meningitis causes secretion of proinflammatory cytokines/chemokines followed by the recruitment of leukocytes into the CNS. In this study, we analyzed the cellular and molecular mechanisms of polymorphonuclear neutrophil (PMN) and monocyte transepithelial transmigration (TM) across the BCSFB after bacterial infection. Methods Using an inverted transwell filter system of human choroid plexus papilloma cells (HIBCPP), we studied leukocyte TM rates, the migration route by immunofluorescence, transmission electron microscopy and focused ion beam/scanning electron microscopy, the secretion of cytokines/chemokines by cytokine bead array and posttranslational modification of the signal regulatory protein (SIRP) α via western blot. Results PMNs showed a significantly increased TM across HIBCPP after infection with wild-type Neisseria meningitidis (MC58). In contrast, a significantly decreased monocyte transmigration rate after bacterial infection of HIBCPP could be observed. Interestingly, in co-culture experiments with PMNs and monocytes, TM of monocytes was significantly enhanced. Analysis of paracellular permeability and transepithelial electrical resistance confirmed an intact barrier function during leukocyte TM. With the help of the different imaging techniques we could provide evidence for para- as well as for transcellular migrating leukocytes. Further analysis of secreted cytokines/chemokines showed a distinct pattern after stimulation and transmigration of PMNs and monocytes. Moreover, the transmembrane glycoprotein SIRPα was deglycosylated in monocytes, but not in PMNs, after bacterial infection. Conclusions Our findings demonstrate that PMNs and monoctyes differentially migrate in a human BCSFB model after bacterial infection. Cytokines and chemokines as well as transmembrane proteins such as SIRPα may be involved in this process.
Collapse
|
25
|
Zhang Z, Chen X, Li L, Zhang K, Tian S, Gao H, Li H. t-PA reduces ischemic impairment of blood-brain barrier by strengthening endothelium junction. Neurol Sci 2013; 34:1605-11. [PMID: 23423463 DOI: 10.1007/s10072-013-1293-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 01/05/2013] [Indexed: 12/11/2022]
Abstract
Cerebral ischemic stroke is one of the most prevalent diseases in senior individuals. Its therapeutical strategies include anticoagulation, thrombolysis and cell protection. Tissue-type plasminogen activator (t-PA) that interacts with thrombin for the lysis of thrombosis is widely used to treat stroke patients in early stage. The mechanism of action of t-PA is not clear. Here, we report a novel role of t-PA in protecting blood-brain barrier and its potential mechanisms. In a model of the blood-brain barrier with human umbilical vascular epithelium cells, we found that t-PA in low concentrations prevented the impairment of the blood-brain barrier as a result of oxygen and glucose deprivation. This protection was fulfilled by strengthening the junctions among vascular endothelia and by upregulating the productions of vascular endothelium growth factor and of zonula occludens-1. Therefore, t-PA may strengthen the junctions of vascular endothelia in the blood-brain barrier to improve the microenvironment of brain cells and, in turn, the outcome of stroke patients.
Collapse
Affiliation(s)
- Zhongling Zhang
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You-Zheng Street, Nangang District, Harbin, 150001, Heilongjiang, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
26
|
Kuckleburg CJ, Newman PJ. Neutrophil proteinase 3 acts on protease-activated receptor-2 to enhance vascular endothelial cell barrier function. Arterioscler Thromb Vasc Biol 2012. [PMID: 23202369 DOI: 10.1161/atvbaha.112.300474] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE The principle role of the vascular endothelium is to present a semi-impermeable barrier to soluble factors and circulating cells, while still permitting the passage of leukocytes from the bloodstream into the tissue. The process of diapedesis involves the selective disruption of endothelial cell junctions, which could compromise vascular integrity. It is therefore somewhat surprising that neutrophil transmigration does not significantly impair endothelial barrier function. We examined whether neutrophils might secrete factors that promote vascular integrity during the latter stages of neutrophil transmigration, in particular, the role of neutrophil serine proteinase 3 (PR3). METHODS AND RESULTS Endothelial cells were treated with PR3 either in its soluble form or in a complex form with cell surface NB1. We observed that PR3 mediated the enhancement of endothelial cell junctional integrity and that this required its proteolytic activity, as well as endothelial cell expression of the protease-activated receptor-2. Importantly, PR3 suppressed the vascular permeability changes and disruption of junctional proteins induced by the action of protease-activated receptor-1 agonists. CONCLUSIONS These findings establish the potential for neutrophil-derived PR3 to play a role in reestablishing vascular integrity after leukocyte transmigration and in protecting endothelial cells from protease-activated receptor-1-induced permeability changes that occur during thrombotic and inflammatory events.
Collapse
Affiliation(s)
- Christopher J Kuckleburg
- Blood Research Institute, BloodCenter of Wisconsin, 8727 Watertown Plank Rd, Milwaukee, WI 53051, USA.
| | | |
Collapse
|
27
|
Block H, Zarbock A. The role of the tec kinase Bruton's tyrosine kinase (Btk) in leukocyte recruitment. Int Rev Immunol 2012; 31:104-18. [PMID: 22449072 DOI: 10.3109/08830185.2012.668982] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Recruitment of leukocytes into inflamed tissue is a key component of the immune system. The activation of integrins on leukocytes is required for their recruitment into the inflamed tissue. Btk is a cytoplasmic nonreceptor tyrosine kinase belonging to the Tec-kinase family. It plays a key role in B-cell development and function, and recently published studies revealed important roles of Btk in myeloid cells. Btk might be activated through a variety of receptors leading to activation of integrins as the pivotal element in leukocyte recruitment. This review focuses on the role of Btk in B-lymphocyte homing and in neutrophil recruitment.
Collapse
Affiliation(s)
- Helena Block
- Department of Anesthesiology and Critical Care Medicine, University of Muenster, Muenster, Germany
| | | |
Collapse
|
28
|
Johnson HL, Chen Y, Jin F, Hanson LM, Gamez JD, Pirko I, Johnson AJ. CD8 T cell-initiated blood-brain barrier disruption is independent of neutrophil support. THE JOURNAL OF IMMUNOLOGY 2012; 189:1937-45. [PMID: 22772449 DOI: 10.4049/jimmunol.1200658] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Blood-brain barrier (BBB) disruption is a common feature of numerous neurologic disorders. A fundamental question in these diseases is the extent inflammatory immune cells contribute to CNS vascular permeability. We have previously shown that CD8 T cells play a critical role in initiating BBB disruption in the peptide-induced fatal syndrome model developed by our laboratory. However, myelomonocytic cells such as neutrophils have also been implicated in promoting CNS vascular permeability and functional deficit in murine models of neuroinflammatory disease. For this reason, we evaluated neutrophil depletion in a murine model of CD8 T cell-initiated BBB disruption by employing traditionally used anti-granulocyte receptor-1 mAb RB6-8C5 and Ly-6G-specific mAb 1A8. We report that CNS-infiltrating antiviral CD8 T cells express high levels of granulocyte receptor-1 protein and are depleted by treatment with RB6-8C5. Mice treated with RB6-8C5, but not 1A8, display: 1) intact BBB tight junction proteins; 2) reduced CNS vascular permeability visible by gadolinium-enhanced T1-weighted magnetic resonance imaging; and 3) preservation of motor function. These studies demonstrate that traditional methods of neutrophil depletion with RB6-8C5 are broadly immune ablating. Our data also provide evidence that CD8 T cells initiate disruption of BBB tight junction proteins and CNS vascular permeability in the absence of neutrophil support.
Collapse
Affiliation(s)
- Holly L Johnson
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Mierke CT. Endothelial cell's biomechanical properties are regulated by invasive cancer cells. MOLECULAR BIOSYSTEMS 2012; 8:1639-49. [PMID: 22498801 DOI: 10.1039/c2mb25024a] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Most cancer-related deaths are caused by the ability of cancer cells to metastasize. This process includes the dissemination of cancer cells from the primary tumor side and their migration to targeted organ sites. During the migration of cancer cells through the connective tissue microenvironment, which consists of endothelial cells and extracellular matrix components, biomechanical properties are crucial for the efficiency and speed of cancer cell invasion and subsequently, metastases formation. Biomechanics can enable cancer cells to migrate through tissue, transmigrate through basement membranes as well as endothelial monolayers and form metastases in targeted organs. The current focus of cancer research still lies on the investigation of cancer cell's biochemical and molecular capabilities such as molecular genetics and gene signaling, but these approaches ignore the mechanical nature of the invasion process of cancer cells. Moreover, even the role of the endothelium during the transmigration and invasion of cells is not clear, it has been seen as a passive barrier, but this could not explain all novel findings. This review discusses how cancer cells alter the structural, biochemical and mechanical properties of the endothelium to regulate their own invasiveness through extracellular matrices and hence, through the tissue microenvironment. Finally, this review sheds light on the mechanical properties of cancer cells and the interacting endothelium and points out the importance of the mechanical properties as a critical determinant for the efficiency of cancer cell invasion and the overall progression of cancer. In conclusion, the regulation of the endothelial cell's biomechanical properties by cancer cells is a critical determinant of cancer cell invasiveness and may affect the future development of new cancer treatments.
Collapse
Affiliation(s)
- Claudia T Mierke
- Faculty of Physics and Earth Science, Institute of Experimental Physics I, Soft Matter Physics Division, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
30
|
Huang RB, Eniola-Adefeso O. Shear stress modulation of IL-1β-induced E-selectin expression in human endothelial cells. PLoS One 2012; 7:e31874. [PMID: 22384091 PMCID: PMC3286450 DOI: 10.1371/journal.pone.0031874] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 01/13/2012] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Endothelial cells (ECs) are continuously exposed to hemodynamic forces imparted by blood flow. While it is known that endothelial behavior can be influenced by cytokine activation or fluid shear, the combined effects of these two independent agonists have yet to be fully elucidated. METHODOLOGY We investigated EC response to long-term inflammatory cues under physiologically relevant shear conditions via E-selectin expression where monolayers of human umbilical vein ECs were simultaneously exposed to laminar fluid shear and interleukin-1ß (shear-cytokine activation) in a parallel plate flow chamber. RESULTS AND CONCLUSION Naïve ECs exposed to shear-cytokine activation display significantly higher E-selectin expression for up to 24 hr relative to ECs activated in static (static-cytokine). Peak E-selectin expression occurred after 8-12 hr of continuous shear-cytokine activation contrary to the commonly observed 4-6 hr peak expression in ECs exposed to static-cytokine activation. Cells with some history of high shear conditioning exhibited either high or muted E-selectin expression depending on the durations of the shear pre-conditioning and the ensuing shear-cytokine activation. Overall, the presented data suggest that a high laminar shear enhances acute EC response to interleukin-1ß in naïve or shear-conditioned ECs as may be found in the pathological setting of ischemia/reperfusion injury while conferring rapid E-selectin downregulation to protect against chronic inflammation.
Collapse
Affiliation(s)
- Ryan B. Huang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
31
|
Mierke CT. The biomechanical properties of 3d extracellular matrices and embedded cells regulate the invasiveness of cancer cells. Cell Biochem Biophys 2012; 61:217-36. [PMID: 21516307 DOI: 10.1007/s12013-011-9193-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The malignancy of tumors depends on the biomechanical properties of cancer cells and their microenvironment, which enable cancer cells to migrate through the connective tissue, transmigrate through basement membranes and endothelial monolayers and form metastases in targeted organs. The current focus of cancer research is still based on biological capabilities such as molecular genetics and gene signaling, but these approaches ignore the mechanical nature of the invasion process of cancer cells. This review will focus on how structural, biochemical and mechanical properties of extracellular matrices (ECMs), and adjacent cells regulate the invasiveness of cancer cells. In addition, it presents how cancer cells create their own microenvironment by restructuring of the ECM and by interaction with stromal cells, which then further contribute to the progression of cancer disease. Finally, this review will point out that mechanical properties are a critical determinant for the efficiency of cancer cell invasion and the progression of cancer which might affect the future development of new cancer treatments.
Collapse
Affiliation(s)
- Claudia T Mierke
- Faculty of Physics and Earth Science, Institute of Experimental Physics I, Soft Matter Physics Division, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
32
|
Assimakopoulos SF, Papageorgiou I, Charonis A. Enterocytes’ tight junctions: From molecules to diseases. World J Gastrointest Pathophysiol 2011; 2:123-37. [PMID: 22184542 PMCID: PMC3241743 DOI: 10.4291/wjgp.v2.i6.123] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 08/26/2011] [Accepted: 10/31/2011] [Indexed: 02/06/2023] Open
Abstract
Tight junctions (TJs) are structures between cells where cells appear in the closest possible contact. They are responsible for sealing compartments when epithelial sheets are generated. They regulate the permeability of ions, (macro) molecules and cells via the paracellular pathway. Their structure at the electron microscopic level has been well known since the 1970s; however, only recently has their macromolecular composition been revealed. This review first examines the major macromolecular components of the TJs (occludin, claudins, junctional adhesion molecule and tricellulin) and then the associated macromolecules at the intracellular plaque [zonula occludens (ZO)-1, ZO-2, ZO-3, AF-6, cingulin, 7H6]. Emphasis is given to their interactions in order to begin to understand the mode of assembly of TJs. The functional significance of TJs is detailed and several mechanisms and factors involved are discussed briefly. Emphasis is given to the role of intestinal TJs and the alterations observed or speculated in diverse disease states. Specifically, intestinal TJs may exert a pathogenetic role in intestinal (inflammatory bowel disease, celiac disease) and extraintestinal diseases (diabetes type 1, food allergies, autoimmune diseases). Additionally, intestinal TJs may be secondarily disrupted during the course of diverse diseases, subsequently allowing the bacterial translocation phenomenon and promoting the systemic inflammatory response, which is often associated with clinical deterioration. The major questions in the field are highlighted.
Collapse
|
33
|
Wang LY, Zhang DL, Zheng JF, Zhang Y, Zhang QD, Liu WH. Apelin-13 passes through the ADMA-damaged endothelial barrier and acts on vascular smooth muscle cells. Peptides 2011; 32:2436-43. [PMID: 22001227 DOI: 10.1016/j.peptides.2011.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 10/01/2011] [Accepted: 10/02/2011] [Indexed: 12/26/2022]
Abstract
Asymmetric dimethylarginine (ADMA), an endogenous nitric oxide synthase inhibitor, is associated with vascular dysfunction. The polypeptide apelin mediates two major actions on blood vessels. However, their combined effects on vascular function are not fully understood. The present study aimed to determine the effect of apelin-13 on myosin light chain (MLC) phosphorylation in vascular smooth muscle cells (VSMCs) under ADMA-induced endothelial leakage conditions. To assess the increased permeability induced by ADMA, human umbilical vein endothelium cells (HUVECs) were plated in transwell dishes. The FITC-dextran flux and FITC-apelin-13 flux through the endothelial monolayer were measured. To examine the effect of leakage of apelin-13 on MLC phosphorylation in HUVSMCs, transwell dishes were used to establish a coculture system with HUVECs in upper chambers and HUVSMCs in lower chambers. Western blot was performed to assess the phospho-MLC levels. ADMA increased endothelial permeability in a concentration- and time-dependent manner, accompanied by actin stress fiber assembly and intercellular gap formation. When HUVECs were treated with ADMA, the permeability to both macromolecular dextran and micromolecular apelin-13 increased significantly. Both p38 MAPK inhibitor and NADPH oxidase inhibitor could prevent HUVECs from the increased permeability, and the changes of cytoskeleton and intercellular junction, which were induced by ADMA. Apelin-13 passed through the ADMA-stimulated endothelial monolayer and increased the expression of phospho-MLC in VSMCs. These results suggest that ADMA increases endothelial permeability, which may involve the p38 MAPK and NADPH oxidase pathway. Apelin-13 can pass through the damaged endothelial barrier, and acts directly on VSMCs to increase MLC phosphorylation.
Collapse
Affiliation(s)
- Li-Yan Wang
- Department of Nephrology, Affiliated Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing 100050, China
| | | | | | | | | | | |
Collapse
|
34
|
Issekutz AC, Rowter D, Macmillan HF. Intravenous immunoglobulin G (IVIG) inhibits IL-1- and TNF-α-dependent, but not chemotactic-factor-stimulated, neutrophil transendothelial migration. Clin Immunol 2011; 141:187-96. [PMID: 21917526 DOI: 10.1016/j.clim.2011.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 08/04/2011] [Accepted: 08/05/2011] [Indexed: 11/26/2022]
Abstract
High-dose intravenous immunoglobulin (IVIG) has anti-inflammatory effects via incompletely understood mechanisms. By investigating whether IVIG might modulate neutrophil (PMN) recruitment, we observed that IVIG dose-dependently inhibited (by 30-50%) PMN transendothelial migration (TEM) across human umbilical vein endothelial cells (EC) stimulated with IL-1α, IL-1β, TNF-α or IL-1β+TNF-α. Inhibition required the presence of IVIG with the responding PMNs, was attributable to the F(ab)(2) portion and was unrelated to putative contaminants in IVIG. IVIG did not inhibit IL-1β- or TNF-α-induced increase of PMN adhesion to EC, nor did it affect C5a- or IL-8-induced PMN TEM across unstimulated EC. Effects of IVIG and F(ab)(2) fragments were not associated with PMN activation, assessed by CD62L shedding, CD11b upregulation or PMN shape. Thus, IVIG selectively inhibits PMN TEM across inflammatory-cytokine-stimulated - but not unstimulated - EC, perhaps contributing to therapeutic benefit in chronic inflammation with minimal impact on chemotactic-factor-induced PMN recruitment during acute infection.
Collapse
Affiliation(s)
- Andrew C Issekutz
- Department of Pediatrics, Dalhousie University, IWK Health Centre, Halifax, Nova Scotia, Canada.
| | | | | |
Collapse
|
35
|
Interdigitated microelectrode-based microchip for electrical impedance spectroscopic study of oral cancer cells. Biomed Microdevices 2011; 13:1075-88. [DOI: 10.1007/s10544-011-9577-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
36
|
Untucht C, Rasch J, Fuchs E, Rohde M, Bergmann S, Steinert M. An optimized in vitro blood-brain barrier model reveals bidirectional transmigration of African trypanosome strains. MICROBIOLOGY-SGM 2011; 157:2933-2941. [PMID: 21737496 DOI: 10.1099/mic.0.049106-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The transmigration of African trypanosomes across the human blood-brain barrier (BBB) is the critical step during the course of human African trypanosomiasis. The parasites Trypanosoma brucei gambiense and T. b. rhodesiense are transmitted to humans during the bite of tsetse flies. Trypanosomes multiply within the bloodstream and finally invade the central nervous system (CNS), which leads to the death of untreated patients. This project focused on the mechanisms of trypanosomal traversal across the BBB. In order to establish a suitable in vitro BBB model for parasite transmigration, different human cell lines were used, including ECV304, HBMEC and HUVEC, as well as C6 rat astrocytes. Validation of the BBB models with Escherichia coli HB101 and E. coli K1 revealed that a combination of ECV304 cells seeded on Matrigel as a semi-synthetic basement membrane and C6 astrocytes resulted in an optimal BBB model system. The BBB model showed selective permeability for the pathogenic E. coli K1 strain, and African trypanosomes were able to traverse the optimized ECV304-C6 BBB efficiently. Furthermore, coincubation indicated that paracellular macrophage transmigration does not facilitate trypanosomal BBB traversal. An inverse assembly of the BBB model demonstrated that trypanosomes were also able to transmigrate the optimized ECV304-C6 BBB backwards, indicating the relevance of the CNS as a possible reservoir of a relapsing parasitaemia.
Collapse
Affiliation(s)
- Christopher Untucht
- Institut für Mikrobiologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Janine Rasch
- Institut für Mikrobiologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Elena Fuchs
- Institut für Mikrobiologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Manfred Rohde
- Helmholtz Zentrum für Infektionsforschung (HZI), Medizinische Mikrobiologie, Braunschweig, Germany
| | - Simone Bergmann
- Institut für Mikrobiologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Michael Steinert
- Institut für Mikrobiologie, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
37
|
Transcellular migration of neutrophil granulocytes through the blood-cerebrospinal fluid barrier after infection with Streptococcus suis. J Neuroinflammation 2011; 8:51. [PMID: 21592385 PMCID: PMC3120695 DOI: 10.1186/1742-2094-8-51] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Accepted: 05/18/2011] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND A critical point during the course of bacterial meningitis is the excessive influx of polymorphnuclear neutrophils (PMNs) from the blood into the brain. Both paracellular and transcellular routes of leukocyte transmigration through the blood-brain barrier have been described in CNS diseases so far. Thus, we investigated the mechanism of PMN transmigration through the blood-CSF barrier under inflammatory conditions. METHODS In an "inverted" Transwell culture model of the blood-CSF barrier, the zoonotic agent Streptococcus suis (S. suis) was used to stimulate porcine choroid plexus epithelial cells (PCPECs) specifically from the physiologically relevant basolateral side. Barrier function was analyzed by measuring TEER and TR-dextran-flux, and tight junction morphology was investigated by immunofluorescence. Route and mechanism of PMN transmigration were determined by immunofluorescence, electron microscopy and FACS analysis. Quantitative real time-PCR was used to determine expression levels of ICAM-1 and VCAM-1. RESULTS Here, we show that the transmigration of PMNs through PCPECs was significantly higher after stimulation with TNFα or infection with S. suis strain 10 compared to its non-encapsulated mutant. Barrier function was not significantly affected by PMN migration alone, but in combination with S. suis infection. Tight junction and cytoskeletal actin reorganisation were also observed after stimulation with S. suis or TNFα. Most strikingly, PMNs preferentially migrated across PCPECs via the transcellular route. Extensive sequential analyses of the PMN transmigration process with Apotome(®)-imaging and electron microscopy revealed that paracellular migrating PMNs stop just before tight junctions. Interestingly, PMNs subsequently appeared to proceed by transcellular migration via funnel-like structures developing from the apical membrane. It is noteworthy that some PMNs contained bacteria during the transmigration process. Flow cytometric and transmigration inhibition studies with integrin-specific antibodies showed that PMN traversal is dependent on CD11b/CD18. Analysis of cell adhesion molecules in PCPECs revealed a significant increase of ICAM-1 and VCAM-1 expression after TNFα and S. suis stimulation. CONCLUSION Our data underline the relevance of the blood-CSF barrier as a gate for leukocyte entry into the CNS and suggest a novel transcellular migration step during the pathogenesis of bacterial meningitis.
Collapse
|
38
|
Grassl GA, Faustmann M, Gill N, Zbytnuik L, Merkens H, So L, Rossi FM, McNagny KM, Finlay BB. CD34 mediates intestinal inflammation in Salmonella-infected mice. Cell Microbiol 2011; 12:1562-75. [PMID: 20497179 DOI: 10.1111/j.1462-5822.2010.01488.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
CD34 is a highly glycosylated sialomucin expressed on a variety of cells, ranging from vascular endothelial cells to haematopoietic stem cells. Depending on its glycosylation state, CD34 has been shown to promote or inhibit cell adhesion and migration; however, a functional role for CD34 in the gut has not been determined. Using a model of Salmonella-induced gastroenteritis, we investigated the role of CD34 in the context of infection. Upon oral infection, the number of CD34+ cells detected in the submucosa, vascular endothelium and lamina propria significantly increased in S. Typhimurium-infected C57Bl/6 mice. The pathology of S. Typhimurium-infected C57Bl/6 mice was characterized by recruitment of neutrophils to the site of inflammation, submucosal oedema and crypt destruction. In contrast, Cd34(-/-) mice showed a delayed pathology, a defect in inflammatory cell migration into the intestinal tissue and enhanced survival. Importantly, this was not due to a lack of chemotactic signals in Cd34(-/-) mice as these mice had either similar or significantly higher levels of pro-inflammatory cytokines and chemokines post infection when compared with infected C57/Bl6 control mice. In summary, we demonstrate a novel role for CD34 in enhancing migration of inflammatory cells and thereby exacerbating host-mediated immunopathology in the intestine of S. Typhimurium-infected mice.
Collapse
Affiliation(s)
- Guntram A Grassl
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zarbock A, Ley K. Protein tyrosine kinases in neutrophil activation and recruitment. Arch Biochem Biophys 2011; 510:112-9. [PMID: 21338576 DOI: 10.1016/j.abb.2011.02.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 02/03/2011] [Accepted: 02/10/2011] [Indexed: 12/30/2022]
Abstract
Migration of leukocytes into tissue is a key element of innate and adaptive immunity. The first contact of leukocytes with endothelial cells is mediated by engagement of selectins with their counter-receptors which results in leukocyte rolling. During rolling, leukocytes collect different inflammatory signals that activate intracellular signaling pathways. Integration of these signals induces leukocyte activation, firm arrest, post-adhesion strengthening, intravascular crawling, and transmigration. In neutrophils, like in T-cells and platelets, both G-protein-coupled receptor-dependent and -independent activation pathways exist that lead to integrin activation. Accumulating evidence suggests that different protein tyrosine kinases play key roles in signal transduction pathways regulating neutrophil activation and recruitment to inflammatory sites. This review focuses on the role of protein tyrosine kinases of the Src, Syk, and Tec families for neutrophil activation and recruitment.
Collapse
Affiliation(s)
- Alexander Zarbock
- Department of Anesthesiology and Intensive Care Medicine, University of Münster, Germany.
| | | |
Collapse
|
40
|
Ko YP, Liang X, Smith CW, Degen JL, Höök M. Binding of Efb from Staphylococcus aureus to fibrinogen blocks neutrophil adherence. J Biol Chem 2011; 286:9865-74. [PMID: 21247890 DOI: 10.1074/jbc.m110.199687] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
In addition to its pivotal role in hemostasis, fibrinogen (Fg) and provisional fibrin matrices play important roles in inflammation and regulate innate immune responses by interacting with leukocytes. Efb (the extracellular fibrinogen-binding protein) is a secreted Staphylococcus aureus protein that engages host Fg and complement C3. However, the molecular details underlying the Efb-Fg interaction and the biological relevance of this interaction have not been determined. In the present study, we characterize the interaction of Efb with Fg. We demonstrate that the Fg binding activity is located within the intrinsically disordered N-terminal half of Efb (Efb-N) and that the D fragment of Fg is the region that mediates Efb-N binding. More detailed studies of the Efb-N-Fg interactions using ELISA and surface plasmon resonance analyses revealed that Efb-N exhibits a much higher affinity for Fg than typically observed with Fg-binding MSCRAMMs (microbial surface components recognizing adhesive matrix molecules), and data obtained from ELISA analyses using truncated Efb-N constructs demonstrate that Efb-N contains two binding sites located within residues 30-67 and 68-98, respectively. Efb-N inhibits neutrophil adhesion to immobilized Fg by binding to Fg and blocking the interaction of the protein with the leukocyte integrin receptor, α(M)β(2). A motif in the Fg γ chain previously shown to be central to the α(M)β(2) interaction was shown to be functionally distinguishable from the Efb-N binding site, suggesting that the Fg-Efb interaction indirectly impedes Fg engagement by α(M)β(2). Taken together, these studies provide insights into how Efb interacts with Fg and suggest that Efb may support bacterial virulence at least in part by impeding Fg-driven leukocyte adhesion events.
Collapse
Affiliation(s)
- Ya-Ping Ko
- Center for Infectious and Inflammatory Disease, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
41
|
Lam FW, Burns AR, Smith CW, Rumbaut RE. Platelets enhance neutrophil transendothelial migration via P-selectin glycoprotein ligand-1. Am J Physiol Heart Circ Physiol 2010; 300:H468-75. [PMID: 21169400 DOI: 10.1152/ajpheart.00491.2010] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Platelets are increasingly recognized as important for inflammation in addition to thrombosis. Platelets promote the adhesion of neutrophils [polymorphonuclear neutrophils (PMNs)] to the endothelium; P-selectin and P-selectin glycoprotein ligand (PSGL)-1 have been suggested to participate in these interactions. Whether platelets also promote PMN transmigration across the endothelium is less clear. We tested the hypothesis that platelets enhance PMN transmigration across the inflamed endothelium and that PSGL-1 is involved. We studied the effects of platelets on PMN transmigration in vivo and in vitro using a well-characterized corneal injury model in C57BL/6 mice and IL-1β-stimulated human umbilical vein endothelial cells (HUVECs) under static and dynamic conditions. In vivo, platelet depletion altered PMN emigration from limbal microvessels after injury, with decreased emigration 6 and 12 h after injury. Both PSGL-1-/- and P-selectin-/- mice, but not Mac-1-/- mice, also had reduced PMN emigration at 12 h after injury relative to wild-type control mice. In the in vitro HUVEC model, platelets enhanced PMN transendothelial migration under static and dynamic conditions independent of firm adhesion. Anti-PSGL-1 antibodies markedly inhibited platelet-PMN aggregates, as assessed by flow cytometry, and attenuated the effect of platelets on PMN transmigration under static conditions without affecting firm adhesion. These data support the notion that platelets enhance neutrophil transmigration across the inflamed endothelium both in vivo and in vitro, via a PSGL-1-dependent mechanism.
Collapse
Affiliation(s)
- Fong W Lam
- Department of Pediatrics, Baylor College of Medicine, 1100 Bates, Rm. 6014, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
42
|
Hong J, Kandasamy K, Marimuthu M, Choi CS, Kim S. Electrical cell-substrate impedance sensing as a non-invasive tool for cancer cell study. Analyst 2010; 136:237-45. [PMID: 20963234 DOI: 10.1039/c0an00560f] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cell-substrate interactions are investigated in a number of studies for drug targets including angiogenesis, arteriosclerosis, chronic inflammatory diseases and carcinogenesis. One characteristic of malignant cancerous cells is their ability to invade tissue. Cell adhesion and cytoskeletal activity have served as valuable indicators for understanding the cancer cell behaviours, such as proliferation, migration and invasion. This review focuses on bio-impedance based measurement for monitoring the behaviours in real time and without using labels. Electric cell-substrate impedance sensing (ECIS) provides rich information about cell-substrate interactions, cell-cell communication and cell adhesion. High sensitivity of the ECIS method allows for observing events down to single-cell level and achieving nanoscale resolution of cell-substrate distances. Recently, its miniaturization and integration with fluorescent detection techniques have been highlighted as a new tool to deliver a high-content platform for anticancer drug development.
Collapse
Affiliation(s)
- Jongin Hong
- Department of Chemistry and Institute of Biomedical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ United Kingdom.
| | | | | | | | | |
Collapse
|
43
|
von Wedel-Parlow M, Schrot S, Lemmen J, Treeratanapiboon L, Wegener J, Galla HJ. Neutrophils cross the BBB primarily on transcellular pathways: an in vitro study. Brain Res 2010; 1367:62-76. [PMID: 20875807 DOI: 10.1016/j.brainres.2010.09.076] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Accepted: 09/21/2010] [Indexed: 01/13/2023]
Abstract
The cerebral microcapillary endothelium forms a highly important barrier between the blood and the interstitial fluid of the brain (blood-brain barrier) that controls the passage of molecules and cells in and out of the CNS. Several CNS diseases include leukocyte extravasation through the endothelium via two mechanistically distinct routes, the paracellular and the transcellular pathway. We established a new in vitro model of the inflamed blood-brain barrier consisting of primary cultured porcine brain capillary endothelial cells which express a tight endothelial barrier even under inflammatory conditions. By means of this specialized blood-brain barrier model we extensively studied the transmigration of neutrophils. Electron and scanning force microscopy as well as immunofluorescence imaging captured the penetrating neutrophil on the endothelial cellular body in between the junctions clearly suggesting a transcellular migration pathway. Electric cell-substrate impedance sensing and transendothelial electrical resistance measurements in combination with expression analysis of tight junction proteins demonstrate that the neutrophil-endothelial interaction does not disrupt the barrier. In conclusion, this study, based on an in vitro model of the blood-brain barrier under inflammatory conditions, evidently implicates that neutrophils preferentially migrate across the BBB via the transcellular route without impairing endothelial barrier function whereas paracellular transmigration plays only a minor role if the barrier is strongly expressed.
Collapse
Affiliation(s)
- Magdalena von Wedel-Parlow
- Institut für Biochemie, Westfälische Wilhelms-Universität Münster, Wilhelm-Klemm-Straße 2, D-48149 Münster, Germany
| | | | | | | | | | | |
Collapse
|
44
|
Chung S, Sudo R, Vickerman V, Zervantonakis IK, Kamm RD. Microfluidic platforms for studies of angiogenesis, cell migration, and cell-cell interactions. Sixth International Bio-Fluid Mechanics Symposium and Workshop March 28-30, 2008 Pasadena, California. Ann Biomed Eng 2010; 38:1164-77. [PMID: 20336839 DOI: 10.1007/s10439-010-9899-3] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Recent advances in microfluidic technologies have opened the door for creating more realistic in vitro cell culture methods that replicate many aspects of the true in vivo microenvironment. These new designs (i) provide enormous flexibility in controlling the critical biochemical and biomechanical factors that influence cell behavior, (ii) allow for the introduction of multiple cell types in a single system, (iii) provide for the establishment of biochemical gradients in two- or three-dimensional geometries, and (iv) allow for high quality, time-lapse imaging. Here, some of the recent developments are reviewed, with a focus on studies from our own laboratory in three separate areas: angiogenesis, cell migration in the context of tumor cell-endothelial interactions, and liver tissue engineering.
Collapse
Affiliation(s)
- Seok Chung
- School of Mechanical Engineering, Korea University, Seoul, Korea
| | | | | | | | | |
Collapse
|
45
|
Charoenphol P, Huang RB, Eniola-Adefeso O. Potential role of size and hemodynamics in the efficacy of vascular-targeted spherical drug carriers. Biomaterials 2010; 31:1392-402. [PMID: 19954839 DOI: 10.1016/j.biomaterials.2009.11.007] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Accepted: 11/02/2009] [Indexed: 01/09/2023]
Abstract
Targeting of drug carriers to the vascular wall is of interest for localized delivery of therapeutics in many human diseases. Nanometer-sized spherical particles are widely proposed for use as carriers for vascular targeting, yet very little evidence has been presented as to their ability to interact with the vascular wall. Thus, this work focuses on elucidating the effect of particle size along with hemodynamics, blood rheology, and vessel size on the adhesion efficiency of targeted polymeric spheres to inflamed endothelium in vitro via parallel plate flow chamber assays. We find that the binding efficiency of spheres to the endothelium from blood flow generally increased with increasing particle size, wall shear rate and channel height for particle sizes from 100 nm up to 10 microm. However, nano-sized particles showed minimal adhesion to the endothelium from blood flow in horizontal (gravity or anti-gravity direction) and vertical channels on the order of small to medium-sized venules and arteries when compared to micron-sized spheres. Furthermore, adhesion of nanospheres was not enhanced with pulsatility in flow. Overall, the presented data suggests that spheres 2-5 microm in size are optimal for targeting the wall in medium to large vessels relevant in several cardiovascular diseases.
Collapse
Affiliation(s)
- Phapanin Charoenphol
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
46
|
Park G, Masi T, Choi CK, Kim H, Becker JM, Sparer TE. Screening for novel constitutively active CXCR2 mutants and their cellular effects. Methods Enzymol 2010; 485:481-97. [PMID: 21050933 DOI: 10.1016/b978-0-12-381296-4.00026-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chemokines play an important role in inflammatory, developmental, and homeostatic processes. Deregulation of this system results in various diseases including tumorigenesis and cancer metastasis. Deregulation can occur when constitutively active mutant (CAM) chemokine receptors are locked in the "on" position. This can lead to cellular transformation/tumorigenesis. The CXC chemokine receptor 2 (CXCR2) is a G-protein-coupled receptor (GPCR) expressed on neutrophils, some monocytes, endothelial cells, and some epithelial cells. CXCR2 activation with CXC chemokines induces leukocyte migration, trafficking, leukocyte degranulation, cellular differentiation, and angiogenesis. Activation of CXCR2 can lead to cellular transformation. We hypothesized that CAM CXCR2s may play a role in cancer development. In order to identify CXCR2 CAMs, potential mutant CXCR2 receptors were screened using a modified Saccharomyces cerevisiae high-throughput system. S. cerevisiae has been used successfully to identify GPCR/G-protein interactions and autocrine selection for peptide agonists. The CXCR2 CAMs identified from this screen were characterized in mammalian cells. Their ability to transform cells in vitro was shown using foci formation, soft-agar growth, impedance measurement assays, and in vivo tumor growth following hind flank inoculation into mice. Signaling pathways contributing to cellular transformation were identified using luciferase reporter assays. Studying constitutively active GPCRs is an approach to "capturing" pluridimensional GPCRs in a "locked" activation state. In order to address the residues necessary for CXCR2 activation, we used S. cerevisiae for screening novel CAMs and characterized them using mammalian reporter assays.
Collapse
Affiliation(s)
- Giljun Park
- The University of Tennessee, Department of Microbiology, Knoxville, Tennessee, USA
| | | | | | | | | | | |
Collapse
|
47
|
Minagar A, Long A, Ma T, Jackson TH, Kelley RE, Ostanin DV, Sasaki M, Warren AC, Jawahar A, Cappell B, Alexander JS. Interferon (IFN)-ß1a and IFN-ß1b Block IFN-?-Induced Disintegration of Endothelial Junction Integrity and Barrier. ACTA ACUST UNITED AC 2009; 10:299-307. [PMID: 14741845 DOI: 10.1080/10623320390272299] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Recent clinical trials indicate the efficacy of interferon (IFN)-beta 1b in reducing relapse rate in relapsing-remitting multiple sclerosis (MS), whereas a surge of IFN-gamma precedes and provokes acute relapses. Disruption of the cerebral endothelial barrier and transendothelial migration of inflammatory cell migration into the brain play a significant role in pathogenesis of MS and may be driven by this surge in IFN-gamma. However, the molecular mechanisms underlying the beneficial effects of IFN-beta 1b against the deleterious effects of IFN-gamma on the barrier formed by the junctional proteins remain to be characterized. The authors investigated the effects of IFN-beta 1b, IFN-beta 1a, and IFN-gamma on the integrity of two endothelial junctional proteins, occludin and vascular endothelial-cadherin (VE-cadherin). Human umbilical vein endothelial cell (HUVEC) layers were treated with IFN-beta 1b, IFN-beta 1a, IFN-gamma, IFN-beta 1b plus IFN-gamma, or IFN-beta 1a plus IFN-gamma. IFN-beta 1b, IFN-beta 1a, and IFN-gamma effects on occludin and VE-cadherin integrity and electrical resistance were assessed by Western blotting and immunofluorescence. IFN-gamma significantly reduced occludin expression and produced gaps in endothelial monolayers. VE-cadherin expression was decreased to a lesser extent in endothelial cells exposed to IFN-gamma. IFN-beta 1b significantly attenuated the IFN-gamma-induced decrease in occludin and VE-cadherin expression. The protective effects of IFN-beta 1a on IFN-gamma-treated endothelial cells were similar to those of IFN-beta 1b. IFN-gamma also significantly reduced endothelial monolayer electrical resistance; this effect was blocked by either IFN-beta 1a or IFN-beta 1b. IFN-beta 1a and IFN-beta 1b effectively prevent the IFN-gamma-induced disintegration of the endothelial tight junctions and sustain barrier against the effects of IFN-gamma. The protective effects of IFN-beta on occludin and VE-cadherin stability appear to represent molecular mechanisms for the therapeutic effects of the IFN-beta on blood brain barrier in MS.
Collapse
Affiliation(s)
- A Minagar
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Application of proteomics to neutrophil biology. J Proteomics 2009; 73:552-61. [PMID: 19580889 DOI: 10.1016/j.jprot.2009.06.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 06/23/2009] [Accepted: 06/28/2009] [Indexed: 01/13/2023]
Abstract
Polymorphonuclear leukocytes or neutrophils are a primary effector cell of the innate immune system and contribute to the development of adaptive immunity. Neutrophils participate in both the initiation and resolution of inflammatory responses through a series of highly coordinated molecular and phenotypic changes. To accomplish these changes, neutrophils express numerous receptors and use multiple overlapping and redundant signal transduction pathways. Dysregulation of the activation or resolution pathways plays a role in a number of human diseases. A comprehensive understanding of the regulation of neutrophil responses can be provided by high throughput proteomic technologies and sophisticated computational analysis. The first steps in the application of proteomics to understanding neutrophil biology have been taken. Here we review the application of expression, structural, and functional proteomic studies to neutrophils. Although defining the complex molecular events associated with neutrophil activation is in the early stages, the data generated to date suggest that proteomic technologies will dramatically enhance our understanding of neutrophil biology.
Collapse
|
49
|
Man S, Tucky B, Bagheri N, Li X, Kochar R, Ransohoff RM. alpha4 Integrin/FN-CS1 mediated leukocyte adhesion to brain microvascular endothelial cells under flow conditions. J Neuroimmunol 2009; 210:92-9. [PMID: 19345424 DOI: 10.1016/j.jneuroim.2009.03.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Revised: 03/04/2009] [Accepted: 03/06/2009] [Indexed: 12/12/2022]
Abstract
Insights into sequential leukocyte-endothelial interactions during leukocyte trafficking have been obtained through experiments using human umbilical vein endothelial cells (HUVEC) under flow conditions. To investigate leukocyte-brain endothelial cell interactions, we developed a dynamic in vitro system, using Transfected Human Brain Microvascular Endothelial Cells (THBMEC) and a parallel plate flow chamber. Human peripheral blood mononuclear cells (PBMC) were perfused across confluent THBMEC cultures at a velocity that approximates the rate found in human brain capillaries. Leukocyte-THBMEC interactions were visualized by phase-contrast microscopy, and images were captured on a CCD camera. To simulate inflammatory conditions, we activated THBMEC with the inflammatory cytokines tumor necrosis factor alpha (TNF-alpha) and interferon gamma (IFN-gamma), which up-regulated chemokine and adhesion molecule expression in THBMEC without affecting the distribution of immunoreactivity for tight junction-associated proteins. PBMC adhesion was enhanced by cytokine-mediated activation of THBMEC. G protein-coupled receptor (GPCR) activation was essential for leukocyte-THBMEC interaction, as pertussis toxin (PTX) treatment of PBMC abrogated PBMC adhesion to activated THBMEC. The anti-alpha4 integrin antibody, natalizumab, infused into MS patients, significantly reduced the adhesion of their ex vivo PBMC to activated THBMEC under flow conditions. Further study showed that alternatively spliced fibronectin containing the CS1 region (FN-CS1), but not Vascular Cell Adhesion Molecule type 1 (VCAM-1), was the ligand of alpha4 integrin on activated THBMEC. Blocking FN-CS1 abrogated PBMC adhesion on activated THBMEC, while anti-VCAM-1 antibodies had no effect. These results established a novel in vitro dynamic BBB model. We also demonstrated the dependence of leukocyte-endothelial interactions in this model on alpha4 integrins and FN-CS1.
Collapse
Affiliation(s)
- Shumei Man
- Neuroinflammation Research Center, Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, OH 44195, USA
| | | | | | | | | | | |
Collapse
|
50
|
English AE, Moy AB, Kruse KL, Ward RC, Kirkpatrick SS, Goldman MH. Instrumental noise estimates stabilize and quantify endothelial cell micro-impedance barrier function parameter estimates. Biomed Signal Process Control 2009. [DOI: 10.1016/j.bspc.2008.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|