1
|
Chakraborty S, Tabrizi Z, Bhatt NN, Franciosa SA, Bracko O. A Brief Overview of Neutrophils in Neurological Diseases. Biomolecules 2023; 13:biom13050743. [PMID: 37238612 DOI: 10.3390/biom13050743] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/21/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
Neutrophils are the most abundant leukocyte in circulation and are the first line of defense after an infection or injury. Neutrophils have a broad spectrum of functions, including phagocytosis of microorganisms, the release of pro-inflammatory cytokines and chemokines, oxidative burst, and the formation of neutrophil extracellular traps. Traditionally, neutrophils were thought to be most important for acute inflammatory responses, with a short half-life and a more static response to infections and injury. However, this view has changed in recent years showing neutrophil heterogeneity and dynamics, indicating a much more regulated and flexible response. Here we will discuss the role of neutrophils in aging and neurological disorders; specifically, we focus on recent data indicating the impact of neutrophils in chronic inflammatory processes and their contribution to neurological diseases. Lastly, we aim to conclude that reactive neutrophils directly contribute to increased vascular inflammation and age-related diseases.
Collapse
Affiliation(s)
| | - Zeynab Tabrizi
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA
| | | | | | - Oliver Bracko
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA
- Department of Neurology, University of Miami-Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
2
|
Integrin Regulators in Neutrophils. Cells 2022; 11:cells11132025. [PMID: 35805108 PMCID: PMC9266208 DOI: 10.3390/cells11132025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 02/01/2023] Open
Abstract
Neutrophils are the most abundant leukocytes in humans and are critical for innate immunity and inflammation. Integrins are critical for neutrophil functions, especially for their recruitment to sites of inflammation or infections. Integrin conformational changes during activation have been heavily investigated but are still not fully understood. Many regulators, such as talin, Rap1-interacting adaptor molecule (RIAM), Rap1, and kindlin, are critical for integrin activation and might be potential targets for integrin-regulating drugs in treating inflammatory diseases. In this review, we outline integrin activation regulators in neutrophils with a focus on the above critical regulators, as well as newly discovered modulators that are involved in integrin activation.
Collapse
|
3
|
Habibi N, Brown TD, Adu-Berchie K, Christau S, Raymond JE, Mooney DJ, Mitragotri S, Lahann J. Nanoparticle Properties Influence Transendothelial Migration of Monocytes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:5603-5616. [PMID: 35446569 DOI: 10.1021/acs.langmuir.2c00200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nanoparticle-based delivery of therapeutics to the brain has had limited clinical impact due to challenges crossing the blood-brain barrier (BBB). Certain cells, such as monocytes, possess the ability to migrate across the BBB, making them attractive candidates for cell-based brain delivery strategies. In this work, we explore nanoparticle design parameters that impact both monocyte association and monocyte-mediated BBB transport. We use electrohydrodynamic jetting to prepare nanoparticles of varying sizes, compositions, and elasticity to address their impact on uptake by THP-1 monocytes and permeation across the BBB. An in vitro human BBB model is developed using human cerebral microvascular endothelial cells (hCMEC/D3) for the assessment of migration. We compare monocyte uptake of both polymeric and synthetic protein nanoparticles (SPNPs) of various sizes, as well as their effect on cell migration. SPNPs (human serum albumin/HSA or human transferrin/TF) are shown to promote increased monocyte-mediated transport across the BBB over polymeric nanoparticles. TF SPNPs (200 nm) associate readily, with an average uptake of 138 particles/cell. Nanoparticle loading is shown to influence the migration of THP-1 monocytes. The migration of monocytes loaded with 200 nm TF and 200 nm HSA SPNPs was 2.3-fold and 2.1-fold higher than that of an untreated control. RNA-seq analysis after TF SPNP treatment suggests that the upregulation of several migration genes may be implicated in increased monocyte migration (ex. integrin subunits α M and α L). Integrin β 2 chain combines with either integrin subunit α M chain or integrin subunit α L chain to form macrophage antigen 1 and lymphocyte function-associated antigen 1 integrins. Both products play a pivotal role in the transendothelial migration cascade. Our findings highlight the potential of SPNPs as drug and/or gene delivery platforms for monocyte-mediated BBB transport, especially where conventional polymer nanoparticles are ineffective or otherwise not desirable.
Collapse
Affiliation(s)
- Nahal Habibi
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Tyler D Brown
- Wyss Institute of Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, Boston, Massachusetts 02115, United States
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, Massachusetts 02318, United States
| | - Kwasi Adu-Berchie
- Wyss Institute of Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, Boston, Massachusetts 02115, United States
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, Massachusetts 02318, United States
| | - Stephanie Christau
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jeffery E Raymond
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - David J Mooney
- Wyss Institute of Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, Boston, Massachusetts 02115, United States
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, Massachusetts 02318, United States
| | - Samir Mitragotri
- Wyss Institute of Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, Boston, Massachusetts 02115, United States
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, Massachusetts 02318, United States
| | - Joerg Lahann
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Material Science & Engineering, Department of Macromolecular Science & Engineering, and Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
4
|
Mauersberger C, Hinterdobler J, Schunkert H, Kessler T, Sager HB. Where the Action Is-Leukocyte Recruitment in Atherosclerosis. Front Cardiovasc Med 2022; 8:813984. [PMID: 35087886 PMCID: PMC8787128 DOI: 10.3389/fcvm.2021.813984] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is the leading cause of death worldwide and leukocyte recruitment is a key element of this phenomenon, thus allowing immune cells to enter the arterial wall. There, in concert with accumulating lipids, the invading leukocytes trigger a plethora of inflammatory responses which promote the influx of additional leukocytes and lead to the continued growth of atherosclerotic plaques. The recruitment process follows a precise scheme of tethering, rolling, firm arrest, crawling and transmigration and involves multiple cellular and subcellular players. This review aims to provide a comprehensive up-to-date insight into the process of leukocyte recruitment relevant to atherosclerosis, each from the perspective of endothelial cells, monocytes and macrophages, neutrophils, T lymphocytes and platelets. In addition, therapeutic options targeting leukocyte recruitment into atherosclerotic lesions-or potentially arising from the growing body of insights into its precise mechanisms-are highlighted.
Collapse
Affiliation(s)
- Carina Mauersberger
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Julia Hinterdobler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Heribert Schunkert
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
5
|
Zhou F, Zhang F, Zarnitsyna VI, Doudy L, Yuan Z, Li K, McEver RP, Lu H, Zhu C. The kinetics of E-selectin- and P-selectin-induced intermediate activation of integrin αLβ2 on neutrophils. J Cell Sci 2021; 134:271954. [PMID: 34435628 DOI: 10.1242/jcs.258046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 08/19/2021] [Indexed: 01/03/2023] Open
Abstract
Selectins and integrins are key players in the adhesion and signaling cascade that recruits leukocytes to inflamed tissues. Selectin binding induces β2 integrin binding to slow leukocyte rolling. Here, a micropipette was used to characterize neutrophil adhesion to E-selectin and intercellular adhesion molecule-1 (ICAM-1) at room temperature. The time-dependent adhesion frequency displayed two-stage kinetics, with an E-selectin-mediated fast increase to a low plateau followed by a slow increase to a high plateau mediated by intermediate-affinity binding of integrin αLβ2 to ICAM-1. The αLβ2 activation required more than 5 s contact to E-selectin and spleen tyrosine kinase (Syk) activity. A multi-zone channel was used to analyze αLβ2 activation by P-selectin in separate zones of receptors or antibodies, finding an inverse relationship between the rolling velocity on ICAM-1 and P-selectin dose, and a P-selectin dose-dependent change from bent to extended conformations with a closed headpiece that was faster at 37°C than at room temperature. Activation of αLβ2 exhibited different levels of cooperativity and persistent times depending on the strength and duration of selectin stimulation. These results define the precise timing and kinetics of intermediate activation of αLβ2 by E- and P-selectins.
Collapse
Affiliation(s)
- Fangyuan Zhou
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA
| | - Fang Zhang
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA
| | - Veronika I Zarnitsyna
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA
| | - Larissa Doudy
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA
| | - Zhou Yuan
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA
| | - Kaitao Li
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA
| | - Rodger P McEver
- Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Hang Lu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Cheng Zhu
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA
| |
Collapse
|
6
|
Biomechanics of Neutrophil Tethers. Life (Basel) 2021; 11:life11060515. [PMID: 34073130 PMCID: PMC8230032 DOI: 10.3390/life11060515] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022] Open
Abstract
Leukocytes, including neutrophils, propelled by blood flow, can roll on inflamed endothelium using transient bonds between selectins and their ligands, and integrins and their ligands. When such receptor–ligand bonds last long enough, the leukocyte microvilli become extended and eventually form thin, 20 µm long tethers. Tether formation can be observed in blood vessels in vivo and in microfluidic flow chambers. Tethers can also be extracted using micropipette aspiration, biomembrane force probe, optical trap, or atomic force microscopy approaches. Here, we review the biomechanical properties of leukocyte tethers as gleaned from such measurements and discuss the advantages and disadvantages of each approach. We also review and discuss viscoelastic models that describe the dependence of tether formation on time, force, rate of loading, and cell activation. We close by emphasizing the need to combine experimental observations with quantitative models and computer simulations to understand how tether formation is affected by membrane tension, membrane reservoir, and interactions of the membrane with the cytoskeleton.
Collapse
|
7
|
Recent advances in tumor microenvironment-targeted nanomedicine delivery approaches to overcome limitations of immune checkpoint blockade-based immunotherapy. J Control Release 2021; 332:109-126. [DOI: 10.1016/j.jconrel.2021.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/24/2021] [Accepted: 02/04/2021] [Indexed: 02/07/2023]
|
8
|
Schwartz AB, Campos OA, Criado-Hidalgo E, Chien S, del Álamo JC, Lasheras JC, Yeh YT. Elucidating the Biomechanics of Leukocyte Transendothelial Migration by Quantitative Imaging. Front Cell Dev Biol 2021; 9:635263. [PMID: 33855018 PMCID: PMC8039384 DOI: 10.3389/fcell.2021.635263] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/09/2021] [Indexed: 01/13/2023] Open
Abstract
Leukocyte transendothelial migration is crucial for innate immunity and inflammation. Upon tissue damage or infection, leukocytes exit blood vessels by adhering to and probing vascular endothelial cells (VECs), breaching endothelial cell-cell junctions, and transmigrating across the endothelium. Transendothelial migration is a critical rate-limiting step in this process. Thus, leukocytes must quickly identify the most efficient route through VEC monolayers to facilitate a prompt innate immune response. Biomechanics play a decisive role in transendothelial migration, which involves intimate physical contact and force transmission between the leukocytes and the VECs. While quantifying these forces is still challenging, recent advances in imaging, microfabrication, and computation now make it possible to study how cellular forces regulate VEC monolayer integrity, enable efficient pathfinding, and drive leukocyte transmigration. Here we review these recent advances, paying particular attention to leukocyte adhesion to the VEC monolayer, leukocyte probing of endothelial barrier gaps, and transmigration itself. To offer a practical perspective, we will discuss the current views on how biomechanics govern these processes and the force microscopy technologies that have enabled their quantitative analysis, thus contributing to an improved understanding of leukocyte migration in inflammatory diseases.
Collapse
Affiliation(s)
- Amy B. Schwartz
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Obed A. Campos
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Ernesto Criado-Hidalgo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Juan C. del Álamo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Mechanical Engineering, University of Washington, Seattle, WA, United States
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, United States
| | - Juan C. Lasheras
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Yi-Ting Yeh
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
9
|
Gao WJ, Liu JX, Xie Y, Luo P, Liu ZQ, Liu L, Zhou H. Suppression of macrophage migration by down-regulating Src/FAK/P130Cas activation contributed to the anti-inflammatory activity of sinomenine. Pharmacol Res 2021; 167:105513. [PMID: 33617975 DOI: 10.1016/j.phrs.2021.105513] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 01/01/2023]
Abstract
A large number of macrophages in inflamed sites not only amplify the severity of inflammatory responses but also contribute to the deleterious progression of many chronic inflammatory diseases, autoimmune diseases and cancers. Macrophage migration is a prerequisite for their entry into inflammatory sites and their participation of macrophages in the pathologic processes. Inhibition of macrophage migration is therefore a potential anti-inflammatory mechanism. Moreover, alleviation of inflammation also prevents the macrophages infiltration. Sinomenine (SIN) is an alkaloid derived from the Chinese medicinal plant Sinomenium acutum. It has multiple pharmacological effects, including anti-inflammation, immunosuppression, and anti-arthritis. However, its anti-inflammatory molecular mechanisms and effect on macrophage migration are not fully understood. The purpose of this research was to investigate the pharmacological effects and the molecular mechanism of SIN on macrophage migration in vivo and in vitro as well as to elucidate its anti-inflammatory mechanisms associated with macrophage migration. Our results showed that SIN reduced the number of RAW264.7 cells migrating into inflammatory paws and blocked lipopolysaccharide (LPS)-induced RAW264.7 cells and bone marrow-derived macrophages (BMDMs) migration in vitro. Furthermore, SIN attenuated the 3D mesenchymal migration of BMDMs. The absence of macrophage migration after circulatory and periphery macrophages depletion led to a reduction in the severity of inflammatory response. In macrophages depleted (macrophages-/-) mice, as inflammatory severity decreased, RAW264.7 cells migration was suppressed. A non-obvious effect of SIN on the inflammatory response was found in macrophages-/- mice, while the inhibitory effect of SIN on RAW264.7 cells migration was still observed. Furthermore, the migration of RAW264.7 cells pre-treated with SIN was suppressed in normal mice. Finally, Src/focal adhesion kinase (FAK)/P130Cas axis activation, which supports macrophages mesenchymal migration, and iNOS expression, NO production, integrin αV and in integrin β3 expressions, which promote Src/FAK/P130Cas activation, were down-regulated by SIN. However, SIN had no obvious effect on the expression of the monocyte chemoattractant protein-1 (MCP-1), which is an important chemokine for macrophage migration. These results indicated that SIN significantly inhibited macrophage mesenchymal migration by down-regulating on Src/FAK/P130Cas axis activation. There was a mutual regulatory correlation between the inflammatory response and macrophage migration, and the effects of SIN on macrophage migration were involved in its anti-inflammatory activity.
Collapse
Affiliation(s)
- Wan-Jiao Gao
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China
| | - Jian-Xin Liu
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua City, Hunan Province, PR China
| | - Yie Xie
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China
| | - Pei Luo
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China
| | - Zhong-Qiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Liang Liu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China; Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou, PR China.
| | - Hua Zhou
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China; Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou, PR China.
| |
Collapse
|
10
|
Park EJ, Myint PK, Ito A, Appiah MG, Darkwah S, Kawamoto E, Shimaoka M. Integrin-Ligand Interactions in Inflammation, Cancer, and Metabolic Disease: Insights Into the Multifaceted Roles of an Emerging Ligand Irisin. Front Cell Dev Biol 2020; 8:588066. [PMID: 33195249 PMCID: PMC7649757 DOI: 10.3389/fcell.2020.588066] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/05/2020] [Indexed: 01/10/2023] Open
Abstract
Integrins are transmembrane proteins that mediate cellular adhesion and migration to neighboring cells or the extracellular matrix, which is essential for cells to undertake diverse physiological and pathological pathways. For integrin activation and ligand binding, bidirectional signaling across the cell membrane is needed. Integrins aberrantly activated under pathologic conditions facilitate cellular infiltration into tissues, thereby causing inflammatory or tumorigenic progressions. Thus, integrins have emerged to the forefront as promising targets for developing therapeutics to treat autoimmune and cancer diseases. In contrast, it remains a fact that integrin-ligand interactions are beneficial for improving the health status of different tissues. Among these ligands, irisin, a myokine produced mainly by skeletal muscles in an exercise-dependent manner, has been shown to bind to integrin αVβ5, alleviating symptoms under unfavorable conditions. These findings may provide insights into some of the underlying mechanisms by which exercise improves quality of life. This review will discuss the current understanding of integrin-ligand interactions in both health and disease. Likewise, we not only explain how diverse ligands play different roles in mediating cellular functions under both conditions via their interactions with integrins, but also specifically highlight the potential roles of the emerging ligand irisin in inflammation, cancer, and metabolic disease.
Collapse
Affiliation(s)
- Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Phyoe Kyawe Myint
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Atsushi Ito
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Thoracic and Cardiovascular Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Michael G Appiah
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Samuel Darkwah
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
11
|
Choo YW, Kang M, Kim HY, Han J, Kang S, Lee JR, Jeong GJ, Kwon SP, Song SY, Go S, Jung M, Hong J, Kim BS. M1 Macrophage-Derived Nanovesicles Potentiate the Anticancer Efficacy of Immune Checkpoint Inhibitors. ACS NANO 2018; 12:8977-8993. [PMID: 30133260 DOI: 10.1021/acsnano.8b02446] [Citation(s) in RCA: 314] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Cancer immunotherapy modulates immune cells to induce antitumor immune responses. Tumors employ immune checkpoints to evade immune cell attacks. Immune checkpoint inhibitors such as anti-PD-L1 antibody (aPD-L1), which is being used clinically for cancer treatments, can block immune checkpoints so that the immune system can attack tumors. However, immune checkpoint inhibitor therapy may be hampered by polarization of macrophages within the tumor microenvironment (TME) into M2 tumor-associated macrophages (TAMs), which suppress antitumor immune responses and promote tumor growth by releasing anti-inflammatory cytokines and angiogenic factors. In this study, we used exosome-mimetic nanovesicles derived from M1 macrophages (M1NVs) to repolarize M2 TAMs to M1 macrophages that release pro-inflammatory cytokines and induce antitumor immune responses and investigated whether the macrophage repolarization can potentiate the anticancer efficacy of aPD-L1. M1NV treatment induced successful polarization of M2 macrophages to M1 macrophages in vitro and in vivo. Intravenous injection of M1NVs into tumor-bearing mice suppressed tumor growth. Importantly, injection of a combination of M1NVs and aPD-L1 further reduced the tumor size, compared to the injection of either M1NVs or aPD-L1 alone. Thus, our study indicates that M1NV injection can repolarize M2 TAMs to M1 macrophages and potentiate antitumor efficacy of the checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Yeon Woong Choo
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Mikyung Kang
- Interdisciplinary Program for Bioengineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Han Young Kim
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Jin Han
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Seokyung Kang
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Ju-Ro Lee
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Gun-Jae Jeong
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Seuk Young Song
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Seokhyeong Go
- Interdisciplinary Program for Bioengineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
- Interdisciplinary Program for Bioengineering , Seoul National University , Seoul 08826 , Republic of Korea
- Institute of Chemical Processes , Seoul National University , Seoul 08826 , Republic of Korea
| |
Collapse
|
12
|
Qasaimeh MA, Pyzik M, Astolfi M, Vidal SM, Juncker D. Neutrophil Chemotaxis in Moving Gradients. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/adbi.201700243] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Mohammad A. Qasaimeh
- Biomedical Engineering Department; McGill University; Montréal QC H3A 0G1 Canada
- Division of Engineering; New York University Abu Dhabi; Abu Dhabi 129188 UAE
- Department of Mechanical and Aerospace Engineering; New York University; NY 11201 USA
| | - Michal Pyzik
- Department of Human Genetics; McGill University; Montréal QC H3G 0B1 Canada
- Division of Gastroenterology; Department of Medicine; Brigham &Women's Hospital; Harvard Medical School; Boston MA 02115 USA
| | - Mélina Astolfi
- Biomedical Engineering Department; McGill University; Montréal QC H3A 0G1 Canada
| | - Silvia M. Vidal
- Department of Human Genetics; McGill University; Montréal QC H3G 0B1 Canada
| | - David Juncker
- Biomedical Engineering Department; McGill University; Montréal QC H3A 0G1 Canada
- Genome Quebec Innovation Centre; McGill University; Montréal QC H3A 0G1 Canada
- Department of Neurology and Neurosurgery; McGill University; Montréal QC H3A 1A4 Canada
| |
Collapse
|
13
|
Hosseini E, Ghasemzadeh M. Intravascular leukocyte migration through platelet thrombi: directing leukocytes to sites of vascular injury. Thromb Haemost 2017; 113:1224-35. [DOI: 10.1160/th14-08-0662] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 01/13/2015] [Indexed: 12/15/2022]
Abstract
SummaryLeukocytes recruitment to thrombi supports an intimate cellular interaction leading to the enhancement of pro-coagulant functions and pro-inflammatory responses at site of vascular injury. Recent observations of neutrophil extracellular traps (NETs) formation and its mutual reactions with platelet thrombi adds more clinical interest to the growing body of knowledge in the field of platelet-leukocyte crosstalk. However, having considered thrombus as a barrier between leukocytes and injured endothelium, the full inflammatory roles of these cells during thrombosis is still ill defined. The most recent observation of neutrophils migration into the thrombi is a phenomenon that highlights the inflammatory functions of leukocytes at the site of injury. It has been hypothesised that leukocytes migration might be associated with the conveyance of highly reactive pro-inflammatory and/or procoagulant mediators to sites of vascular injury. In addition, the evidence of neutrophils migration into arterial thrombi following traumatic and ischaemia-reperfusion injury highlights the already described role of these cells in atherosclerosis. Regardless of the mechanisms behind leukocyte migration, whether these migrated cells benefit normal homeostasis by their involvement in wound healing and vascular rebuilding or they increase unwilling inflammatory responses, could be of interest for future researches that provide new insight into biological importance of leukocyte recruitment to thrombi.
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW Since the discovery of the lack of kindlin-3 expression as the reason for the immunopathology leukocyte adhesion deficiency III syndrome, the role of kindlin-3 in inflammatory processes was investigated in a numerous studies. This review gives an overview about recent findings regarding the role of kindlin-3 in neutrophil activation and recruitment. RECENT FINDINGS Kindlin-3, together with talin-1, contributes essentially to the activation of β2-integrins in neutrophils. During inside-out signaling, kindlin-3 binds to the β-cytoplasmic integrin tail and is indispensable for the integrin conformational shift into the high-affinity ligand binding conformation, but not for the intermediate (extended) conformation. During outside-in signaling (as a consequence of integrin ligand binding) kindlin-3 interacts with distinct signaling molecules and is required for cell-autonomous functions like migration and spreading. SUMMARY Leukocyte adhesion deficiency III syndrome, which is caused by absence of kindlin-3, is a rarely occurring disease. However, the investigation of the clinical symptoms as well as the underlying molecular mechanisms gave rise to a huge amount of new insights into the processes of integrin activation in neutrophils and the consequences of defects in these processes.
Collapse
|
15
|
Selectin catch-bonds mechanotransduce integrin activation and neutrophil arrest on inflamed endothelium under shear flow. Blood 2017; 130:2101-2110. [PMID: 28811304 DOI: 10.1182/blood-2017-05-783027] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/03/2017] [Indexed: 12/27/2022] Open
Abstract
E-selectin extends from the plasma membrane of inflamed endothelium and serves to capture leukocytes from flowing blood via long-lived catch-bonds that support slow leukocyte rolling under shear stress. Its ligands are glycosylated with the tetrasaccharide sialyl Lewisx (sLex), which contributes to bond affinity and specificity. E-selectin-mediated rolling transmits signals into neutrophils that trigger activation of high-affinity β2-integrins necessary for transition to shear-resistant adhesion and transendothelial migration. Rivipansel is a glycomimetic drug that inhibits E-selectin-mediated vaso-occlusion induced by integrin-dependent sickle-red blood cell-leukocyte adhesion. How Rivipansel antagonizes ligand recognition by E-selectin and blocks outside-in signaling of integrin-mediated neutrophil arrest while maintaining rolling immune-surveillance is unknown. Here, we demonstrate that sLex expressed on human L-selectin is preferentially bound by E-selectin and, on ligation, initiates secretion of MRP8/14 that binds TLR4 to elicit the extension of β2-integrin to an intermediate affinity state. Neutrophil rolling over E-selectin at precise shear stress transmits tension and catch-bond formation with L-selectin via sLex, resulting in focal clusters that deliver a distinct signal to upshift β2-integrins to a high-affinity state. Rivipansel effectively blocked formation of selectin catch-bonds, revealing a novel mechanotransduction circuit that rapidly converts extended β2-integrins to high-affinity shear-resistant bond clusters with intracellular adhesion molecule 1 on inflamed endothelium.
Collapse
|
16
|
Fan Z, Ley K. Leukocyte arrest: Biomechanics and molecular mechanisms of β2 integrin activation. Biorheology 2016; 52:353-77. [PMID: 26684674 DOI: 10.3233/bir-15085] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Integrins are a group of heterodimeric transmembrane receptors that play essential roles in cell-cell and cell-matrix interaction. Integrins are important in many physiological processes and diseases. Integrins acquire affinity to their ligand by undergoing molecular conformational changes called activation. Here we review the molecular biomechanics during conformational changes of integrins, integrin functions in leukocyte biorheology (adhesive functions during rolling and arrest) and molecules involved in integrin activation.
Collapse
Affiliation(s)
- Zhichao Fan
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.,Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
17
|
Gao L, Gülcüler GS, Golbach L, Block H, Zarbock A, Martin-Villalba A. Endothelial cell-derived CD95 ligand serves as a chemokine in induction of neutrophil slow rolling and adhesion. eLife 2016; 5. [PMID: 27763263 PMCID: PMC5098908 DOI: 10.7554/elife.18542] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 10/19/2016] [Indexed: 12/31/2022] Open
Abstract
Integrin activation is crucial for the regulation of leukocyte rolling, adhesion and trans-vessel migration during inflammation and occurs by engagement of myeloid cells through factors presented by inflamed vessels. However, endothelial-dependent mechanisms of myeloid cell recruitment are not fully understood. Here we show using an autoperfused flow chamber assay of whole blood neutrophils and intravital microscopy of the inflamed cremaster muscle that CD95 mediates leukocyte slow rolling, adhesion and transmigration upon binding of CD95-ligand (CD95L) that is presented by endothelial cells. In myeloid cells, CD95 triggers activation of Syk-Btk/PLCγ2/Rap1 signaling that ultimately leads to integrin activation. Excitingly, CD95-deficient myeloid cells exhibit impaired bacterial clearance in an animal model of sepsis induced by cecal ligation and puncture (CLP). Our data identify the cellular and molecular mechanisms underlying the chemoattractant effect of endothelial cell-derived CD95L in induction of neutrophil recruitment and support the use of therapeutic inhibition of CD95’s activity in inflammatory diseases. DOI:http://dx.doi.org/10.7554/eLife.18542.001 When tissues are damaged or infected, the body produces an inflammatory response. Neutrophils – a type of white blood cell – play an important part in this response. These cells normally circulate through the bloodstream, and are recruited to the inflamed site by chemical signals sent out by immune cells in the damaged tissue. This causes passing neutrophils to migrate through the wall of the blood vessel to gain access to the inflamed tissue. The neutrophils go through a sequence of steps before they can pass through the blood vessel wall. After initially tethering to the cells that line the blood vessel, the neutrophils experience a period of “slow rolling” across the vessel lining, before tightly adhering to one of the cells. In 2010, researchers determined that a protein on the neutrophil’s surface, known as CD95, helps the cell migrate through blood vessel walls. This protein interacts with a “ligand” molecule on the surface of the cells that line the blood vessel. However, it remains unclear whether CD95 and its ligand play a role in the steps that lead up to the neutrophils migrating through the blood vessel wall. Gao et al. – who include researchers involved in the 2010 study – now show that activating CD95 in neutrophils also triggers the cell’s slow rolling and adhesion. Experiments performed on mouse cells and tissues showed that the cells that line the blood vessels present the CD95 ligand on their surfaces in order to activate CD95 in the neutrophils circulating in the bloodstream. This ultimately leads to neutrophil slow rolling and adhesion. Further experiments in mice showed that this ability of CD95 to recruit neutrophils to inflamed sites was crucial for clearing bacteria in cases of sepsis, where infection causes the immune system to damage the body’s own tissues. Future studies could address whether inhibiting CD95's activity could help to treat diseases that feature uncontrolled white blood cell recruitment, including various cancers and autoimmune diseases. DOI:http://dx.doi.org/10.7554/eLife.18542.002
Collapse
Affiliation(s)
- Liang Gao
- Division of Molecular Neurobiology, German Cancer Research Center, Heidelberg, Germany
| | - Gülce Sila Gülcüler
- Division of Molecular Neurobiology, German Cancer Research Center, Heidelberg, Germany
| | - Lieke Golbach
- Department of Anesthesiology and Critical Care Medicine, University of Münster, Münster, Germany
| | - Helena Block
- Department of Anesthesiology and Critical Care Medicine, University of Münster, Münster, Germany
| | - Alexander Zarbock
- Department of Anesthesiology and Critical Care Medicine, University of Münster, Münster, Germany
| | - Ana Martin-Villalba
- Division of Molecular Neurobiology, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
18
|
Pertussis Toxin Exploits Host Cell Signaling Pathways Induced by Meningitis-Causing E. coli K1-RS218 and Enhances Adherence of Monocytic THP-1 Cells to Human Cerebral Endothelial Cells. Toxins (Basel) 2016; 8:toxins8100291. [PMID: 27754355 PMCID: PMC5086651 DOI: 10.3390/toxins8100291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 09/30/2016] [Accepted: 10/01/2016] [Indexed: 01/13/2023] Open
Abstract
Pertussis toxin (PTx), the major virulence factor of the whooping cough-causing bacterial pathogen Bordetella pertussis, permeabilizes the blood–brain barrier (BBB) in vitro and in vivo. Breaking barriers might promote translocation of meningitis-causing bacteria across the BBB, thereby facilitating infection. PTx activates several host cell signaling pathways exploited by the neonatal meningitis-causing Escherichia coli K1-RS218 for invasion and translocation across the BBB. Here, we investigated whether PTx and E. coli K1-RS218 exert similar effects on MAPK p38, NF-κB activation and transcription of downstream targets in human cerebral endothelial TY10 cells using qRT-PCR, Western blotting, and ELISA in combination with specific inhibitors. PTx and E. coli K1-RS218 activate MAPK p38, but only E. coli K1-RS218 activates the NF-κB pathway. mRNA and protein levels of p38 and NF-κB downstream targets including IL-6, IL-8, CxCL-1, CxCL-2 and ICAM-1 were increased. The p38 specific inhibitor SB203590 blocked PTx-enhanced activity, whereas E. coli K1-RS218’s effects were inhibited by the NF-κB inhibitor Bay 11-7082. Further, we found that PTx enhances the adherence of human monocytic THP-1 cells to human cerebral endothelial TY10 cells, thereby contributing to enhanced translocation. These modulations of host cell signaling pathways by PTx and meningitis-causing E. coli support their contributions to pathogen and monocytic THP-1 cells translocation across the BBB.
Collapse
|
19
|
Schopohl P, Grüneberg P, Melzig M. The influence of harpagoside and harpagide on TNFα-secretion and cell adhesion molecule mRNA-expression in IFNγ/LPS-stimulated THP-1 cells. Fitoterapia 2016; 110:157-65. [DOI: 10.1016/j.fitote.2016.03.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/04/2016] [Accepted: 03/05/2016] [Indexed: 01/13/2023]
|
20
|
Herrmann JM, Meyle J. Neutrophil activation and periodontal tissue injury. Periodontol 2000 2015; 69:111-27. [DOI: 10.1111/prd.12088] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
21
|
Germena G, Volmering S, Sohlbach C, Zarbock A. Mutation in the CD45 inhibitory wedge modulates integrin activation and leukocyte recruitment during inflammation. THE JOURNAL OF IMMUNOLOGY 2014; 194:728-38. [PMID: 25505282 DOI: 10.4049/jimmunol.1401646] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Neutrophil recruitment to the site of inflammation plays a pivotal role in host defense. Src family kinases (SFKs) activation is required for integrin and chemokine signaling as well as immune cell function. The receptor-like protein tyrosine phosphatase CD45 positively regulates chemoattractant signaling acting on SFK activity. To further investigate the role of CD45 in neutrophil recruitment and function, we analyzed transgenic mice carrying a single point mutation (CD45E613R), which constitutively activates CD45. By using intravital microscopy experiments, we demonstrated that different steps of the leukocyte recruitment cascade were affected in CD45E613R mutant mice. The rolling velocity of CD45E613R mutant neutrophils was decreased compared with wild-type neutrophils that subsequently resulted in an increased number of adherent cells. The analysis of β2 integrins LFA-1 and macrophage-1 Ag (Mac-1) showed that in CD45E613R mutant neutrophils LFA-1 adhesiveness was impaired, and avidity was enhanced, whereas Mac-1 adhesiveness was increased. Because of the increased Mac-1 adhesiveness, neutrophil crawling was impaired in CD45E613R mutant compared with wild-type neutrophils. In an Escherichia coli lung infection model, CD45E613R mice displayed a decreased neutrophil recruitment into the alveolar compartment, which resulted in an increased number of CFUs in the lung. Our data demonstrate that the CD45E613R mutation modulates integrin activation and leukocyte recruitment during inflammation.
Collapse
Affiliation(s)
- Giulia Germena
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital of Münster, 48149 Münster, Germany; andMax-Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Stephanie Volmering
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital of Münster, 48149 Münster, Germany; andMax-Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Charlotte Sohlbach
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital of Münster, 48149 Münster, Germany; andMax-Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital of Münster, 48149 Münster, Germany; andMax-Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| |
Collapse
|
22
|
Futosi K, Fodor S, Mócsai A. Reprint of Neutrophil cell surface receptors and their intracellular signal transduction pathways. Int Immunopharmacol 2013; 17:1185-97. [PMID: 24263067 DOI: 10.1016/j.intimp.2013.11.010] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 12/07/2012] [Accepted: 06/09/2013] [Indexed: 12/13/2022]
Abstract
Neutrophils play a critical role in the host defense against bacterial and fungal infections, but their inappropriate activation also contributes to tissue damage during autoimmune and inflammatory diseases. Neutrophils express a large number of cell surface receptors for the recognition of pathogen invasion and the inflammatory environment. Those include G-protein-coupled chemokine and chemoattractant receptors, Fc-receptors, adhesion receptors such as selectins/selectin ligands and integrins, various cytokine receptors, as well as innate immune receptors such as Toll-like receptors and C-type lectins. The various cell surface receptors trigger very diverse signal transduction pathways including activation of heterotrimeric and monomeric G-proteins, receptor-induced and store-operated Ca(2+) signals, protein and lipid kinases, adapter proteins and cytoskeletal rearrangement. Here we provide an overview of the receptors involved in neutrophil activation and the intracellular signal transduction processes they trigger. This knowledge is crucial for understanding how neutrophils participate in antimicrobial host defense and inflammatory tissue damage and may also point to possible future targets of the pharmacological therapy of neutrophil-mediated autoimmune or inflammatory diseases.
Collapse
Affiliation(s)
- Krisztina Futosi
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary
| | | | | |
Collapse
|
23
|
Futosi K, Fodor S, Mócsai A. Neutrophil cell surface receptors and their intracellular signal transduction pathways. Int Immunopharmacol 2013; 17:638-50. [PMID: 23994464 PMCID: PMC3827506 DOI: 10.1016/j.intimp.2013.06.034] [Citation(s) in RCA: 457] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 12/07/2012] [Accepted: 06/09/2013] [Indexed: 12/29/2022]
Abstract
Neutrophils play a critical role in the host defense against bacterial and fungal infections, but their inappropriate activation also contributes to tissue damage during autoimmune and inflammatory diseases. Neutrophils express a large number of cell surface receptors for the recognition of pathogen invasion and the inflammatory environment. Those include G-protein-coupled chemokine and chemoattractant receptors, Fc-receptors, adhesion receptors such as selectins/selectin ligands and integrins, various cytokine receptors, as well as innate immune receptors such as Toll-like receptors and C-type lectins. The various cell surface receptors trigger very diverse signal transduction pathways including activation of heterotrimeric and monomeric G-proteins, receptor-induced and store-operated Ca2 + signals, protein and lipid kinases, adapter proteins and cytoskeletal rearrangement. Here we provide an overview of the receptors involved in neutrophil activation and the intracellular signal transduction processes they trigger. This knowledge is crucial for understanding how neutrophils participate in antimicrobial host defense and inflammatory tissue damage and may also point to possible future targets of the pharmacological therapy of neutrophil-mediated autoimmune or inflammatory diseases. Neutrophils are crucial players in innate and adaptive immunity. Neutrophils also participate in autoimmune and inflammatory diseases. Various neutrophil receptors recognize pathogens and the inflammatory environment. The various cell surface receptors trigger diverse intracellular signaling. Neutrophil receptors and signaling are potential targets in inflammatory diseases.
Collapse
Affiliation(s)
- Krisztina Futosi
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary
| | - Szabina Fodor
- Department of Computer Science, Corvinus University of Budapest, 1093 Budapest, Hungary
| | - Attila Mócsai
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary
- Corresponding author at: Department of Physiology, Semmelweis University School of Medicine, Tűzoltó utca 37–47, 1094 Budapest, Hungary. Tel.: + 36 1 459 1500x60 409; fax: + 36 1 266 7480.
| |
Collapse
|
24
|
Pick R, Brechtefeld D, Walzog B. Intraluminal crawling versus interstitial neutrophil migration during inflammation. Mol Immunol 2013; 55:70-5. [DOI: 10.1016/j.molimm.2012.12.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 11/13/2012] [Accepted: 12/06/2012] [Indexed: 12/23/2022]
|
25
|
Abstract
Quantitative dynamic footprinting (qDF) allows visualization of the footprints of live leukocytes rolling on a selectin-coated cover glass. qDF works on the principle of total internal reflection fluorescence, which involves fluorescence excitation in a thin slice (~200 nm) of the cell proximal to the cover glass while the rest of the cell remains dark. Dual color qDF (DqDF) is an advancement of qDF, which enables simultaneous visualization of two fluorochromes in the footprints of rolling leukocytes. When the fluorochrome is localized either in the cell cytoplasm or plasma membrane, the two-dimensional qDF image is used to create a three-dimensional rendition of the footprint topography. DqDF is a useful tool to study leukocyte adhesion under flow, and has recently been used to reveal mechanisms that enable neutrophils to roll at high shear stresses that prevail in venules during inflammation.
Collapse
Affiliation(s)
- Prithu Sundd
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | |
Collapse
|
26
|
Ghasemzadeh M, Hosseini E. Platelet-leukocyte crosstalk: Linking proinflammatory responses to procoagulant state. Thromb Res 2012; 131:191-7. [PMID: 23260445 DOI: 10.1016/j.thromres.2012.11.028] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 11/20/2012] [Accepted: 11/23/2012] [Indexed: 01/18/2023]
Abstract
Platelet activation is known to be associated with the release of a vast array of chemokines and proinflammatory lipids which induce pleiotropic effects on a wide variety of tissues and cells, including leukocytes. During thrombosis, the recruitment of leukocytes to activated platelets is considered an important step which not only links thrombosis to inflammatory responses but may also enhance procoagulant state. This phenomenon is highly regulated and influenced by precise mutual interactions between the cells at site of vascular injury and thrombi formation. Platelet-leukocyte interaction involves a variety of mediators including adhesion molecules, chemokines and chemoattractant molecules, shed proteins, various proinflammatory lipids and other materials. The current review addresses the detailed mechanisms underlying platelet-leukocyte crosstalk. This includes their adhesive interactions, transcellular metabolisms, induced tissue factor activity and neutrophil extracellular traps formation as well as the impacts of these phenomena in modulation of the proinflammatory and procoagulant functions in a reciprocal manner that enhances the physiological responses.
Collapse
Affiliation(s)
- Mehran Ghasemzadeh
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| | | |
Collapse
|
27
|
Silveira AAA, Dominical VM, Lazarini M, Costa FF, Conran N. Simvastatin abrogates inflamed neutrophil adhesive properties, in association with the inhibition of Mac-1 integrin expression and modulation of Rho kinase activity. Inflamm Res 2012; 62:127-32. [PMID: 23250325 DOI: 10.1007/s00011-012-0579-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/22/2012] [Accepted: 11/26/2012] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVES Leukocytes play a primary role in vascular inflammation, and thus an understanding of the pathways involved in the activation of these cells and means to inhibit their consequent adhesion to the vessel wall is of significant interest. This study aimed to determine whether statins have a direct effect upon neutrophil adhesive properties under inflammatory conditions. METHODS Neutrophils from healthy individuals were subjected to adhesion assays (with fibronectin as ligand) and flow cytometry. RESULTS In the presence of a TNF-α inflammatory stimulus, neutrophils displayed a rapid and substantial enhancement in their adhesive properties that was abrogated by preincubation of cells with simvastatin. Neutrophil surface expression of the Mac-1 integrin subunit, CD11b, was augmented by TNF-α, and this increased expression was also inhibited by simvastatin. TNF-α also induced neutrophil LFA-1 and Mac-1 activation, but this activation was not blocked by simvastatin. Interestingly, while addition of the isoprenoids, geranygerayl pyrophosphate and farnesyl pyrophosphate, to cells did not alter the effect of simvastatin on TNF-α-stimulated adhesion, concurrent incubation of cells with the Rho kinase (ROCK) inhibitor reversed the effects of simvastatin on neutrophil adhesion and CD11b expression. CONCLUSION Simvastatin appears to have direct anti-inflammatory effects in neutrophils that may be mediated by modulation of ROCK activity.
Collapse
|
28
|
Bianchi E, Molteni R, Pardi R, Dubini G. Microfluidics for in vitro biomimetic shear stress-dependent leukocyte adhesion assays. J Biomech 2012. [PMID: 23200903 DOI: 10.1016/j.jbiomech.2012.10.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recruitment of leukocytes from blood to tissues is a multi-step process playing a major role in the activation of inflammatory responses. Tethering and rolling of leukocytes along the vessel wall, followed by arrest and transmigration through the endothelium result from chemoattractant-dependent signals, inducing adhesive and migratory events. Shear forces exerted by the blood flow on leukocytes induce rolling via selectin-mediated interactions with endothelial cells and increase the probability of leukocytes to engage their chemokine receptors, facilitating integrin activation and consequent arrest. Flow-derived shear forces generate mechanical stimuli concurring with biochemical signals in the modulation of leukocyte-endothelial cell interactions. In the last few years, a host of in vitro studies have clarified the biochemical adhesion cascade and the role of shear stress in leukocyte extravasation. The limitation of the static environment in Boyden devices has been overcome both by the use of parallel-plate flow chambers and by custom models mimicking the in vivo conditions, along with widespread microfluidic approaches to in vitro modeling. These devices create an in vitro biomimetic environment where the multi-step transmigration process can be imaged and quantified under mechanical and biochemical controlled conditions, including fluid dynamic settings, channel design, materials and surface coatings. This paper reviews the technological solutions recently proposed to model, observe and quantify leukocyte adhesion behavior under shear flow, with a final survey of high-throughput solutions featuring multiple parallel assays as well as thorough and time-saving statistical interpretation of the experimental results.
Collapse
Affiliation(s)
- Elena Bianchi
- LaBS-Laboratory of Biological Structure Mechanics, Department of Structural Engineering, Politecnico di Milano, Milan, Italy.
| | | | | | | |
Collapse
|
29
|
Sundd P, Pospieszalska MK, Ley K. Neutrophil rolling at high shear: flattening, catch bond behavior, tethers and slings. Mol Immunol 2012; 55:59-69. [PMID: 23141302 DOI: 10.1016/j.molimm.2012.10.025] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 10/15/2012] [Accepted: 10/16/2012] [Indexed: 12/30/2022]
Abstract
Neutrophil recruitment to sites of inflammation involves neutrophil rolling along the inflamed endothelium in the presence of shear stress imposed by blood flow. Neutrophil rolling in post-capillary venules in vivo is primarily mediated by P-selectin on the endothelium binding to P-selectin glycoprotein ligand-1 (PSGL-1) constitutively expressed on neutrophils. Blood flow exerts a hydrodynamic drag on the rolling neutrophil which is partially or fully balanced by the adhesive forces generated in the P-selectin-PSGL-1 bonds. Rolling is the result of rapid formation and dissociation of P-selectin-PSGL-1 bonds at the center and rear of the rolling cell, respectively. Neutrophils roll stably on P-selectin in post-capillary venules in vivo and flow chambers in vitro at wall shear stresses greater than 6 dyn cm(-2). However, the mechanisms that enable neutrophils to roll at such high shear stress are not completely understood. In vitro and in vivo studies have led to the discovery of four potential mechanisms, viz. cell flattening, catch bond behavior, membrane tethers, and slings. Rolling neutrophils undergo flattening at high shear stress, which not only increases the size of the cell footprint but also reduces the hydrodynamic drag experienced by the rolling cell. P-selectin-PSGL-1 bonds behave as catch bonds at small detachment forces and thus become stronger with increasing force. Neutrophils rolling at high shear stress form membrane tethers which can be longer than the cell diameter and promote the survival of P-selectin-PSGL-1 bonds. Finally, neutrophils rolling at high shear stress form 'slings', which act as cell autonomous adhesive substrates and support step-wise peeling. Tethers and slings act together and contribute to the forces balancing the hydrodynamic drag. How the synergy between the four mechanisms leads to stable rolling at high shear stress is an area that needs further investigation.
Collapse
Affiliation(s)
- Prithu Sundd
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
30
|
Dixit N, Kim MH, Rossaint J, Yamayoshi I, Zarbock A, Simon SI. Leukocyte function antigen-1, kindlin-3, and calcium flux orchestrate neutrophil recruitment during inflammation. THE JOURNAL OF IMMUNOLOGY 2012; 189:5954-64. [PMID: 23144497 DOI: 10.4049/jimmunol.1201638] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Neutrophil arrest and migration on inflamed endothelium involves a conformational shift in CD11a/CD18 (leukocyte function antigen-1; LFA-1) to a high-affinity and clustered state that determines the strength and lifetime of bond formation with ICAM-1. Cytoskeletal adapter proteins Kindlin-3 and Talin-1 anchor clustered LFA-1 to the cytoskeleton and facilitate the transition from neutrophil rolling to arrest. We recently reported that tensile force acts on LFA-1 bonds inducing their colocalization with Orai1, the predominant membrane store operated Ca(2+) channel that cooperates with the endoplasmic reticulum to elicit cytosolic flux. Because Kindlin-3 was recently reported to initiate LFA-1 clustering in lymphocytes, we hypothesized that it cooperates with Orai1 and LFA-1 in signaling local Ca(2+) flux necessary for shear-resistant neutrophil arrest. Using microfluidic flow channels combined with total internal reflection fluorescence microscopy, we applied defined shear stress to low- or high-affinity LFA-1 and imaged the spatiotemporal regulation of bond formation with Kindlin-3 recruitment and Ca(2+) influx. Orai1 and Kindlin-3 genes were silenced in neutrophil-like HL-60 cells to assess their respective roles in this process. Kindlin-3 was enriched within focal clusters of high-affinity LFA-1, which promoted physical linkage with Orai1. This macromolecular complex functioned to amplify inside-out Ca(2+) signaling in response to IL-8 stimulation by catalyzing an increased density of Talin-1 and consolidating LFA-1 clusters within sites of contact with ICAM-1. In this manner, neutrophils use focal adhesions as mechanosensors that convert shear stress-mediated tensile force into local bursts of Ca(2+) influx that catalyze cytoskeletal engagement and an adhesion-strengthened migratory phenotype.
Collapse
Affiliation(s)
- Neha Dixit
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
| | | | | | | | | | | |
Collapse
|
31
|
Rossaint J, Berger C, Van Aken H, Scheld HH, Zahn PK, Rukosujew A, Zarbock A. Cardiopulmonary bypass during cardiac surgery modulates systemic inflammation by affecting different steps of the leukocyte recruitment cascade. PLoS One 2012; 7:e45738. [PMID: 23029213 PMCID: PMC3446926 DOI: 10.1371/journal.pone.0045738] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Accepted: 08/24/2012] [Indexed: 12/30/2022] Open
Abstract
Background It is known that the use of a cardiopulmonary bypass (CPB) during cardiac surgery leads to leukocyte activation and may, among other causes, induce organ dysfunction due to increased leukocyte recruitment into different organs. Leukocyte extravasation occurs in a cascade-like fashion, including capturing, rolling, adhesion, and transmigration. However, the molecular mechanisms of increased leukocyte recruitment caused by CPB are not known. This clinical study was undertaken in order to investigate which steps of the leukocyte recruitment cascade are affected by the systemic inflammation during CPB. Methods We investigated the effects of CPB on the different steps of the leukocyte recruitment cascade in whole blood from healthy volunteers (n = 9) and patients undergoing cardiac surgery with the use of cardiopulmonary bypass (n = 7) or in off-pump coronary artery bypass-technique (OPCAB, n = 9) by using flow chamber experiments, transmigration assays, and biochemical analysis. Results CPB abrogated selectin-induced slow leukocyte rolling on E-selectin/ICAM-1 and P-selectin/ICAM-1. In contrast, chemokine-induced arrest and transmigration was significantly increased by CPB. Mechanistically, the abolishment of slow leukocyte rolling was due to disturbances in intracellular signaling with reduced phosphorylation of phospholipase C (PLC) γ2, Akt, and p38 MAP kinase. Furthermore, CPB induced an elevated transmigration which was caused by upregulation of Mac-1 on neutrophils. Conclusion These data suggest that CPB abrogates selectin-mediated slow leukocyte rolling by disturbing intracellular signaling, but that the clinically observed increased leukocyte recruitment caused by CPB is due to increased chemokine-induced arrest and transmigration. A better understanding of the underlying molecular mechanisms causing systemic inflammation after CPB may aid in the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Christian Berger
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Hugo Van Aken
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Hans H. Scheld
- Department of Thoracic and Cardiovascular Surgery, University Hospital Münster, Münster, Germany
| | - Peter K. Zahn
- Department of Anesthesiology, Intensive Care Medicine, Palliative Care Medicine and Pain Management, University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Andreas Rukosujew
- Department of Thoracic and Cardiovascular Surgery, University Hospital Münster, Münster, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
- * E-mail:
| |
Collapse
|
32
|
Rillahan CD, Antonopoulos A, Lefort CT, Sonon R, Azadi P, Ley K, Dell A, Haslam SM, Paulson JC. Global metabolic inhibitors of sialyl- and fucosyltransferases remodel the glycome. Nat Chem Biol 2012; 8:661-8. [PMID: 22683610 PMCID: PMC3427410 DOI: 10.1038/nchembio.999] [Citation(s) in RCA: 325] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 05/05/2012] [Indexed: 12/23/2022]
Abstract
Despite the fundamental roles of sialyl- and fucosyltransferases in mammalian physiology, there are few pharmacological tools to manipulate their function in a cellular setting. Although fluorinated analogs of the donor substrates are well-established transition state inhibitors of these enzymes, they are not membrane permeable. By exploiting promiscuous monosaccharide salvage pathways, we show that fluorinated analogs of sialic acid and fucose can be taken up and metabolized to the desired donor substrate-based inhibitors inside the cell. Because of the existence of metabolic feedback loops, they also act to prevent the de novo synthesis of the natural substrates, resulting in a global, family-wide shutdown of sialyl- and/or fucosyltransferases and remodeling of cell-surface glycans. As an example of the functional consequences, the inhibitors substantially reduce expression of the sialylated and fucosylated ligand sialyl Lewis X on myeloid cells, resulting in loss of selectin binding and impaired leukocyte rolling.
Collapse
Affiliation(s)
- Cory D. Rillahan
- Department of Chemical Physiology The Scripps Research Institute La Jolla, CA 92037 (USA)
| | - Aristotelis Antonopoulos
- Division of Molecular Biosciences Faculty of Natural Sciences mperial College London, London SW7 2AZ (UK)
| | - Craig T. Lefort
- La Jolla Institute for Allergy and Immunology Division of Inflammation Biology La Jolla, CA 92037 (USA)
| | - Roberto Sonon
- Complex Carbohydrate Research Center The University of Georgia Athens, GA 30602 (USA)
| | - Parastoo Azadi
- Complex Carbohydrate Research Center The University of Georgia Athens, GA 30602 (USA)
| | - Klaus Ley
- La Jolla Institute for Allergy and Immunology Division of Inflammation Biology La Jolla, CA 92037 (USA)
| | - Anne Dell
- Division of Molecular Biosciences Faculty of Natural Sciences mperial College London, London SW7 2AZ (UK)
| | - Stuart M. Haslam
- Division of Molecular Biosciences Faculty of Natural Sciences mperial College London, London SW7 2AZ (UK)
| | - James C. Paulson
- Department of Chemical Physiology The Scripps Research Institute La Jolla, CA 92037 (USA)
| |
Collapse
|
33
|
Abstract
Lymphocyte function-associated antigen-1 (LFA-1) is a heterodimeric integrin consisting of αL (gene name, Itgal) and β2 (gene name, Itgb2) subunits expressed in all leukocytes. LFA-1 is essential for neutrophil recruitment to inflamed tissue. Activation of LFA-1 by chemokines allows neutrophils and other leukocytes to undergo arrest, resulting in firm adhesion on endothelia expressing intercellular adhesion molecules (ICAMs). In mice, CXCR2 is the primary chemokine receptor involved in triggering neutrophil arrest, and it does so through “inside-out” activation of LFA-1. CXCR2 signaling induces changes in LFA-1 conformation that are coupled to affinity upregulation of the ligand-binding headpiece (extended with open I domain). Unlike naïve lymphocytes, engagement of P-selectin glycoprotein ligand-1 (PSGL-1) on neutrophils stimulates a slow rolling behavior that is mediated by LFA-1 in a distinct activation state (extended with closed I domain). How inside-out signaling cascades regulate the structure and function of LFA-1 is being studied using flow chambers, intravital microscopy, and flow cytometry for ligand and reporter antibody binding. Here, we review how LFA-1 activation is regulated by cellular signaling and ligand binding. Two FERM domain-containing proteins, talin-1 and Kindlin-3, are critical integrin co-activators and have distinct roles in the induction of LFA-1 conformational rearrangements. This review integrates these new results into existing models of LFA-1 activation.
Collapse
Affiliation(s)
- Craig T Lefort
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | | |
Collapse
|
34
|
Montresor A, Toffali L, Constantin G, Laudanna C. Chemokines and the signaling modules regulating integrin affinity. Front Immunol 2012; 3:127. [PMID: 22654882 PMCID: PMC3360201 DOI: 10.3389/fimmu.2012.00127] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/03/2012] [Indexed: 01/05/2023] Open
Abstract
Integrin-mediated adhesion is a general concept referring to a series of adhesive phenomena including tethering–rolling, affinity, valency, and binding stabilization altogether controlling cell avidity (adhesiveness) for the substrate. Arrest chemokines modulate each aspect of integrin activation, although integrin affinity regulation has been recognized as the prominent event in rapid leukocyte arrest induced by chemokines. A variety of inside-out and outside-in signaling mechanisms have been related to the process of integrin-mediated adhesion in different cellular models, but only few of them have been clearly contextualized to rapid integrin affinity modulation by arrest chemokines in primary leukocytes. Complex signaling processes triggered by arrest chemokines and controlling leukocyte integrin activation have been described for ras-related rap and for rho-related small GTPases. We summarize the role of rap and rho small GTPases in the regulation of rapid integrin affinity in primary leukocytes and provide a modular view of these pro-adhesive signaling events. A potential, albeit still speculative, mechanism of rho-mediated regulation of cytoskeletal proteins controlling the last step of integrin activation is also discussed. We also discuss data suggesting a functional integration between the rho- and rap-modules of integrin activation. Finally we examine the universality of signaling mechanisms regulating integrin triggering by arrest chemokines.
Collapse
Affiliation(s)
- Alessio Montresor
- Division of General Pathology, Department of Pathology, University of Verona Verona, Italy
| | | | | | | |
Collapse
|
35
|
Lefort CT, Rossaint J, Moser M, Petrich BG, Zarbock A, Monkley SJ, Critchley DR, Ginsberg MH, Fässler R, Ley K. Distinct roles for talin-1 and kindlin-3 in LFA-1 extension and affinity regulation. Blood 2012; 119:4275-82. [PMID: 22431571 PMCID: PMC3359742 DOI: 10.1182/blood-2011-08-373118] [Citation(s) in RCA: 184] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 03/09/2012] [Indexed: 01/07/2023] Open
Abstract
In inflammation, neutrophils and other leukocytes roll along the microvascular endothelium before arresting and transmigrating into inflamed tissues. Arrest requires conformational activation of the integrin lymphocyte function-associated antigen-1 (LFA-1). Mutations of the FERMT3 gene encoding kindlin-3 underlie the human immune deficiency known as leukocyte adhesion deficiency-III. Both kindlin-3 and talin-1, another FERM domain-containing cytoskeletal protein, are required for integrin activation, but their individual roles in the induction of specific integrin conformers are unclear. Here, we induce differential LFA-1 activation in neutrophils through engagement of the selectin ligand P-selectin glycoprotein ligand-1 or the chemokine receptor CXCR2. We find that talin-1 is required for inducing LFA-1 extension, which corresponds to intermediate affinity and induces neutrophil slow rolling, whereas both talin-1 and kindlin-3 are required for induction of the high-affinity conformation of LFA-1 with an open headpiece, which results in neutrophil arrest. In vivo, both slow rolling and arrest are defective in talin-1-deficient neutrophils, whereas only arrest is defective in kindlin-3-deficient neutrophils. We conclude that talin-1 and kindlin-3 serve distinct functions in LFA-1 activation.
Collapse
Affiliation(s)
- Craig T Lefort
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease and ulcerative colitis, is associated with enhanced leukocyte infiltration to the gut, which is directly linked to the clinical aspects of these disorders. Thus, leukocyte trafficking is a major target for IBD therapy. Past and emerging techniques to study leukocyte trafficking both in vitro and in vivo have expanded our knowledge of the leukocyte migration process and the role of inhibitors. Various strategies have been employed to target chemokine- and integrin-ligand interactions within the multistep adhesion cascade and the S1P/S1PR1 axis in leukocyte migration. Though there is an abundance of preclinical data demonstrating efficacy of leukocyte trafficking inhibitors, many have yet to be confirmed in clinical studies. Vigilance for toxicity and further research is required into this complex and emerging area of IBD therapy.
Collapse
|
37
|
Sundd P, Pospieszalska MK, Cheung LSL, Konstantopoulos K, Ley K. Biomechanics of leukocyte rolling. Biorheology 2011; 48:1-35. [PMID: 21515934 DOI: 10.3233/bir-2011-0579] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Leukocyte rolling on endothelial cells and other P-selectin substrates is mediated by P-selectin binding to P-selectin glycoprotein ligand-1 expressed on the tips of leukocyte microvilli. Leukocyte rolling is a result of rapid, yet balanced formation and dissociation of selectin-ligand bonds in the presence of hydrodynamic shear forces. The hydrodynamic forces acting on the bonds may either increase (catch bonds) or decrease (slip bonds) their lifetimes. The force-dependent 'catch-slip' bond kinetics are explained using the 'two pathway model' for bond dissociation. Both the 'sliding-rebinding' and the 'allosteric' mechanisms attribute 'catch-slip' bond behavior to the force-induced conformational changes in the lectin-EGF domain hinge of selectins. Below a threshold shear stress, selectins cannot mediate rolling. This 'shear-threshold' phenomenon is a consequence of shear-enhanced tethering and catch bond-enhanced rolling. Quantitative dynamic footprinting microscopy has revealed that leukocytes rolling at venular shear stresses (>0.6 Pa) undergo cellular deformation (large footprint) and form long tethers. The hydrodynamic shear force and torque acting on the rolling cell are thought to be synergistically balanced by the forces acting on tethers and stressed microvilli, however, their relative contribution remains to be determined. Thus, improvement beyond the current understanding requires in silico models that can predict both cellular and microvillus deformation and experiments that allow measurement of forces acting on individual microvilli and tethers.
Collapse
Affiliation(s)
- Prithu Sundd
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| | | | | | | | | |
Collapse
|
38
|
Abstract
Progenitor cells mobilized from the bone marrow are recruited to ischemic tissues and increase neovascularization. Cell therapy is a promising new therapeutic option for treating patients with ischemic disorders. The efficiency of cell therapy to augment recovery after ischemia depends on the sufficient recruitment and engraftment of the cells to the target tissue. Homing to sites of active neovascularization is a complex process depending on a timely and spatially orchestrated interplay between chemokines, chemokine receptors, adhesion molecules (selectins and integrins), and intracellular signaling cascades, including also oxidative signaling. This review will focus on the homing mechanisms of progenitor and stem cells to ischemic tissues. Specifically, we discuss the role of chemokines and adhesion molecules such as selectins and integrins and the crosstalk between chemokines and integrins in progenitor cell homing.
Collapse
|
39
|
Stadtmann A, Brinkhaus L, Mueller H, Rossaint J, Bolomini-Vittori M, Bergmeier W, Van Aken H, Wagner DD, Laudanna C, Ley K, Zarbock A. Rap1a activation by CalDAG-GEFI and p38 MAPK is involved in E-selectin-dependent slow leukocyte rolling. Eur J Immunol 2011; 41:2074-85. [PMID: 21480213 DOI: 10.1002/eji.201041196] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 03/19/2011] [Accepted: 04/04/2011] [Indexed: 12/14/2022]
Abstract
Rolling leukocytes are exposed to different adhesion molecules and chemokines. Neutrophils rolling on E-selectin induce integrin αLβ2-mediated slow rolling on ICAM-1 by activating a phospholipase C (PLC)γ2-dependent and a separate PI3Kγ-dependent pathway. E-selectin-signaling cooperates with chemokine signaling to recruit neutrophils into inflamed tissues. However, the distal signaling pathway linking PLCγ2 (Plcg2) to αLβ2-activation is unknown. To identify this pathway, we used different Tat-fusion-mutants and gene-deficient mice in intravital microscopy, autoperfused flow chamber, peritonitis, and biochemical studies. We found that the small GTPase Rap1 is activated following E-selectin engagement and that blocking Rap1a in Pik3cg-/- mice by a dominant-negative Tat-fusion mutant completely abolished E-selectin-mediated slow rolling. We identified CalDAG-GEFI (Rasgrp2) and p38 MAPK as key signaling intermediates between PLCγ2 and Rap1a. Gαi-independent leukocyte adhesion to and transmigration through endothelial cells in inflamed postcapillary venules of the cremaster muscle were completely abolished in Rasgrp2-/- mice. The physiological importance of CalDAG-GEFI in E-selectin-dependent integrin activation is shown by complete inhibition of neutrophil recruitment into the inflamed peritoneal cavity of Rasgrp2-/- leukocytes treated with pertussis toxin to block Gαi-signaling. Our data demonstrate that Rap1a activation by p38 MAPK and CalDAG-GEFI is involved in E-selectin-dependent slow rolling and leukocyte recruitment.
Collapse
Affiliation(s)
- Anika Stadtmann
- Department of Anesthesiology and Intensive Care Medicine, University of Münster, Münster, Germany; Max-Planck Institute Münster, Münster, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Acute loss of renal function attenuates slow leukocyte rolling and transmigration by interfering with intracellular signaling. Kidney Int 2011; 80:493-503. [PMID: 21562471 DOI: 10.1038/ki.2011.125] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Acute loss of renal function reduces leukocyte recruitment into inflamed tissues, and we studied the molecular basis of this using intravital microscopy of cremaster muscle and an autoperfused flow chamber system after bilateral nephrectomy or sham operation in mice. Acute loss of renal function resulted in cessation of selectin-induced slow leukocyte rolling on E-selectin/intercellular adhesion molecule 1 (ICAM-1) and P-selectin/ICAM-1. It also reduced in vivo neutrophil extravasation (assessed by reflected light oblique transillumination) without affecting chemokine-induced arrest. This elimination of selectin-mediated slow leukocyte rolling was associated with a reduced phosphorylation of spleen tyrosine kinase, Akt, phospholipase C-γ2, and p38 MAPK. However, the levels of adhesion molecules located on the neutrophil surface were not altered. Leukocytes from critically ill patients with sepsis-induced acute kidney injury showed a significantly higher rolling velocity on E-selectin/ICAM-1- and P-selectin/ICAM-1-coated surfaces compared with patients with sepsis alone or healthy volunteers. Thus, an acute loss of renal function significantly impairs neutrophil rolling and transmigration, both in vivo and in vitro. These effects are due, in part, to decreased phosphorylation of selectin-dependent intracellular signaling pathways.
Collapse
|
41
|
|
42
|
Laudanna C, Bolomini-Vittori M. Integrin activation in the immune system. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2010; 1:116-127. [PMID: 20835985 DOI: 10.1002/wsbm.9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Modulation of leukocyte adhesiveness is critical to leukocyte function during the immune response. A central paradigm in this phenomenon is represented by integrin activation, which is controlled by inside-out signal transduction mechanisms triggered by selectins, chemoattractants and TcR-bound Ag and facilitated by mechanochemical forces. Integrins are heterodimeric adhesive receptors differently expressed on all leukocyte subtypes. At least two distinct modalities of integrin activation are known, namely conformational changes, leading to increased affinity, and lateral mobility leading to increased valency, both enhancing cell avidity (adhesiveness). Several signal transduction events have been correlated to integrin activation in leukocytes. The complexity of intracellular signaling networks leading to leukocyte integrin activation is likely functional to generate robustness and fine tuning of integrin activation allowing integration of qualitative and quantitative variations of extracellular signals leading to leukocyte-, agonist- and integrin-specific control of adhesion. In this context, the recent modular abstraction proposed for the functional architecture of biological networks may provide a powerful paradigm to understand regulation and specificity of signaling events. Accordingly, pro-adhesive intracellular signaling networks may be organized in regulatory signalosomes, or modules, corresponding to discrete clusters of interacting signaling proteins, with some devoted to context-dependent regulation of specificity and dynamics of integrin activation. The principles and technologies of systems biology, and more specifically of network theory, may help to address this complexity and unveil the inner logic governing leukocyte recruitment during the immune response.
Collapse
Affiliation(s)
- Carlo Laudanna
- Department of Pathology, The Center for Biomedical Computing (CBMC), University of Verona, Verona, Italy
| | - Matteo Bolomini-Vittori
- Department of Pathology, The Center for Biomedical Computing (CBMC), University of Verona, Verona, Italy
| |
Collapse
|
43
|
Canalli AA, Proença RF, Franco-Penteado CF, Traina F, Sakamoto TM, Saad STO, Conran N, Costa FF. Participation of Mac-1, LFA-1 and VLA-4 integrins in the in vitro adhesion of sickle cell disease neutrophils to endothelial layers, and reversal of adhesion by simvastatin. Haematologica 2010; 96:526-33. [PMID: 21173096 DOI: 10.3324/haematol.2010.032912] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Pharmacological approaches to inhibit increased leukocyte adhesive interactions in sickle cell disease may represent important strategies for the prevention of vaso-occlusion in patients with this disorder. We investigated, in vitro, the adhesion molecules involved in endothelial-sickle cell disease neutrophil interactions and the effect of simvastatin on sickle cell disease neutrophil adhesion to tumor necrosis factor-α-activated endothelial monolayers (human umbilical vein endothelial cells), and neutrophil chemotaxis. DESIGN AND METHODS Sickle cell disease patients in steady state and not on hydroxyurea were included in the study. Endothelial cells treated, or not, with tumor necrosis factor-α and simvastatin were used for neutrophil adhesion assays. Neutrophils treated with simvastatin were submitted to interleukin 8-stimulated chemotaxis assays. RESULTS Sickle cell disease neutrophils showed greater adhesion to endothelial cells than control neutrophils. Adhesion of control neutrophils to endothelial cells was mediated by Mac-1 under basal conditions and by the Mac-1 and LFA-1 integrins under inflammatory conditions. In contrast, adhesion of sickle cell disease neutrophils to endothelium, under both basal and tumor necrosis factor-α-stimulated conditions, was mediated by Mac-1 and LFA-1 integrins and also by VLA-4. Under stimulated inflammatory conditions, simvastatin significantly reduced sickle cell disease neutrophil adhesion, and this effect was reversed by inhibition of nitric oxide synthase. Furthermore, intercellular adhesion molecule-1 expression was significantly abrogated on tumor necrosis factor-α-stimulated endothelium incubated with simvastatin, and statin treatment inhibited the interleukin-8-stimulated migration of both control and sickle cell disease neutrophils. CONCLUSIONS The integrins Mac-1, LFA-1 and, interestingly, VLA-4 mediate the adhesion of sickle cell disease leukocytes to activated endothelial cell layers, in vitro. Our data indicate that simvastatin may be able to reduce endothelial activation and consequent leukocyte adhesion in this in vitro model; future experiments and clinical trials may determine whether simvastatin therapy could be employed in patients with sickle cell disease, with beneficial effects on vaso-occlusion.
Collapse
Affiliation(s)
- Andreia A Canalli
- Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Chidlow JH, Glawe JD, Alexander JS, Kevil CG. VEGF₁₆₄ differentially regulates neutrophil and T cell adhesion through ItgaL- and ItgaM-dependent mechanisms. Am J Physiol Gastrointest Liver Physiol 2010; 299:G1361-7. [PMID: 20884890 PMCID: PMC3006242 DOI: 10.1152/ajpgi.00202.2010] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Leukocyte recruitment to inflamed tissues is the cornerstone of inflammatory responses and the driving force behind the establishment of inflammatory bowel disease, consisting of Crohn's disease and ulcerative colitis. It has been reported that angiogenic cytokines contribute to this inflammatory response that facilitates the chronic nature of disease. We have previously reported (Goebel S, Huang M, Davis WC, Jennings M, Siahaan TJ, Alexander JS, Kevil CG. Am J Physiol Gastrointest Liver Physiol 290: G648-G654, 2006) that vascular endothelial growth factor (VEGF)-A can stimulate neutrophil adhesion to colon microvascular endothelial cells in a β₂-integrin (Itgb2)-dependent manner. However, it is not known which of the specific leukocyte integrins are critical for VEGF-A-dependent neutrophil and T cell recruitment. Here we examine the differential importance of either α-integrin (Itga)L or ItgaM in governing neutrophil and T cell adhesion to VEGF-A-activated colonic endothelium. Using an in vitro parallel-plate flow chamber model, we found that genetic deficiency of ItgaM completely blunted neutrophil adhesion to VEGF-A-stimulated endothelium, whereas ItgaL deficiency only partly blocked neutrophil adhesion. Deficiency of ItgaM did significantly decrease neutrophil rolling, whereas deficiency of ItgaL did not. We found that genetic deficiency of either ItgaL or ItgaM did significantly blunt T cell adhesion to VEGF-A-stimulated colon endothelium. We also found that genetic deficiency of these Itgas significantly attenuated T cell rolling behavior. Lastly, we examined whether VEGF-A-mediated leukocyte recruitment occurred through different VEGF receptor (VEGFR) pathways and found that VEGFR2 activation regulates neutrophil recruitment, whereas both VEGFR1 and VEGFR2 modulate T cell recruitment. Together, these data identify differential molecular mechanisms of VEGF-A-mediated leukocyte recruitment.
Collapse
Affiliation(s)
- John H. Chidlow
- 2Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | | | - J. Steven Alexander
- 2Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Christopher G. Kevil
- Departments of 1Pathology and ,2Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| |
Collapse
|
45
|
Quantitative dynamic footprinting microscopy reveals mechanisms of neutrophil rolling. Nat Methods 2010; 7:821-4. [PMID: 20871617 PMCID: PMC2967732 DOI: 10.1038/nmeth.1508] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Accepted: 08/31/2010] [Indexed: 01/13/2023]
Abstract
We introduce quantitative dynamic footprinting microscopy to resolve neutrophil rolling on P-selectin. We show that the footprint of a rolling neutrophil is four times larger than previously thought, the P-selectin-PSGL-1 bonds are relaxed at the leading edge of the rolling cell, compressed under the cell center, and stretched at the trailing edge. Each rolling neutrophil also forms 3-4 long tethers that extend up to 16 μm behind the rolling cell.
Collapse
|
46
|
Schaff UY, Trott KA, Chase S, Tam K, Johns JL, Carlyon JA, Genetos DC, Walker NJ, Simon SI, Borjesson DL. Neutrophils exposed to A. phagocytophilum under shear stress fail to fully activate, polarize, and transmigrate across inflamed endothelium. Am J Physiol Cell Physiol 2010; 299:C87-96. [PMID: 20392928 PMCID: PMC2904253 DOI: 10.1152/ajpcell.00165.2009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Accepted: 04/02/2010] [Indexed: 11/22/2022]
Abstract
Anaplasma phagocytophilum is an obligate intracellular bacterium that has evolved mechanisms to hijack polymorphonuclear neutrophil (PMN) receptors and signaling pathways to bind, infect, and multiply within the host cell. E-selectin is upregulated during inflammation and is a requisite endothelial receptor that supports PMN capture, rolling, and activation of integrin-mediated arrest. Ligands expressed by PMN that mediate binding to endothelium via E-selectin include sialyl Lewis x (sLe(x))-expressing ligands such as P-selectin glycoprotein ligand-1 (PSGL-1) and other glycolipids and glycoproteins. As A. phagocytophilum is capable of binding to sLe(x)-expressing ligands expressed on PMN, we hypothesized that acute bacterial adhesion to PMN would subsequently attenuate PMN recruitment during inflammation. We assessed the dynamics of PMN recruitment and migration under shear flow in the presence of a wild-type strain of A. phagocytophilum and compared it with a strain of bacteria that binds to PMN independent of PSGL-1. Acute bacterial engagement with PMN resulted in transient PMN arrest and minimal PMN polarization. Although the wild-type pathogen also signaled activation of beta2 integrins and elicited a mild intracellular calcium flux, downstream signals including PMN transmigration and phosphorylation of p38 mitogen-activated protein kinase (MAPK) were inhibited. The mutant strain bound less well to PMN and failed to activate beta2 integrins and induce a calcium flux but did result in decreased PMN arrest and polarization that may have been partially mediated by a suppression of p38 MAPK activation. This model suggests that A. phagocytophilum binding to PMN under shear flow during recruitment to inflamed endothelium interferes with normal tethering via E-selectin and navigational signaling of transendothelial migration.
Collapse
Affiliation(s)
- U Y Schaff
- Department of Biomedical Engineering, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Tang J, Hunt CA. Identifying the rules of engagement enabling leukocyte rolling, activation, and adhesion. PLoS Comput Biol 2010; 6:e1000681. [PMID: 20174606 PMCID: PMC2824748 DOI: 10.1371/journal.pcbi.1000681] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Accepted: 01/20/2010] [Indexed: 11/18/2022] Open
Abstract
The LFA-1 integrin plays a pivotal role in sustained leukocyte adhesion to the endothelial surface, which is a precondition for leukocyte recruitment into inflammation sites. Strong correlative evidence implicates LFA-1 clustering as being essential for sustained adhesion, and it may also facilitate rebinding events with its ligand ICAM-1. We cannot challenge those hypotheses directly because it is infeasible to measure either process during leukocyte adhesion following rolling. The alternative approach undertaken was to challenge the hypothesized mechanisms by experimenting on validated, working counterparts: simulations in which diffusible, LFA1 objects on the surfaces of quasi-autonomous leukocytes interact with simulated, diffusible, ICAM1 objects on endothelial surfaces during simulated adhesion following rolling. We used object-oriented, agent-based methods to build and execute multi-level, multi-attribute analogues of leukocytes and endothelial surfaces. Validation was achieved across different experimental conditions, in vitro, ex vivo, and in vivo, at both the individual cell and population levels. Because those mechanisms exhibit all of the characteristics of biological mechanisms, they can stand as a concrete, working theory about detailed events occurring at the leukocyte-surface interface during leukocyte rolling and adhesion experiments. We challenged mechanistic hypotheses by conducting experiments in which the consequences of multiple mechanistic events were tracked. We quantified rebinding events between individual components under different conditions, and the role of LFA1 clustering in sustaining leukocyte-surface adhesion and in improving adhesion efficiency. Early during simulations ICAM1 rebinding (to LFA1) but not LFA1 rebinding (to ICAM1) was enhanced by clustering. Later, clustering caused both types of rebinding events to increase. We discovered that clustering was not necessary to achieve adhesion as long as LFA1 and ICAM1 object densities were above a critical level. Importantly, at low densities LFA1 clustering enabled improved efficiency: adhesion exhibited measurable, cell level positive cooperativity.
Collapse
Affiliation(s)
- Jonathan Tang
- UCSF/UC Berkeley Joint Graduate Group in Bioengineering, University of California, Berkeley, Berkeley, California, United States of America
| | - C. Anthony Hunt
- UCSF/UC Berkeley Joint Graduate Group in Bioengineering, University of California, Berkeley, Berkeley, California, United States of America
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
48
|
Abstract
Neutrophil recruitment into inflamed tissue in response to injury or infection is tightly regulated. Reduced neutrophil recruitment can result in a reduced ability to fight invading microorganisms. During inflammation, neutrophils roll along the endothelial wall of postcapillary venules and integrate inflammatory signals. Neutrophil activation by selectins and chemokines regulates integrin adhesiveness. Binding of activated integrins to their counter-receptors on endothelial cells induces neutrophil arrest and firm adhesion. Adherent neutrophils can be further activated to undergo cytoskeletal rearrangement, crawling, transmigration, superoxide production, and respiratory burst. Signaling through G-protein-coupled receptors, selectin ligands, Fc receptors and outside-in signaling through integrins are all involved in neutrophil activation, but their interplay in the multistep process of recruitment is only beginning to emerge. This review provides an overview of signaling in rolling and adherent neutrophils.
Collapse
Affiliation(s)
- Alexander Zarbock
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | |
Collapse
|
49
|
Pospieszalska MK, Ley K. Chapter 8 Modeling Leukocyte Rolling. CURRENT TOPICS IN MEMBRANES 2009. [DOI: 10.1016/s1063-5823(09)64008-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
50
|
Zarbock A, Ley K. New insights into leukocyte recruitment by intravital microscopy. Curr Top Microbiol Immunol 2009; 334:129-52. [PMID: 19521684 DOI: 10.1007/978-3-540-93864-4_6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Leukocyte recruitment to sites of inflammation requires adhesion to and transmigration through the blood vessel wall. Recent progress in optical equipment and new genetic and molecular tools have revealed additional steps in the leukocyte adhesion cascade beyond rolling, adhesion, and transmigration. In vivo studies using intravital microscopy (IVM) were essential for the discovery of slow rolling, postadhesion strengthening, intraluminal crawling, and different routes of transmigration. IVM revealed unique features of leukocyte recruitment in different organs. This review focuses on insights into the leukocyte adhesion cascade gained by IVM.
Collapse
Affiliation(s)
- Alexander Zarbock
- Department of Anesthesiology and Intensive Care Medicine, University of Münster, Münster, Germany.
| | | |
Collapse
|