1
|
Nguyen H, Podolnikova NP, Ugarova TP, Wang X. α MI-domain of integrin Mac-1 binds the cytokine pleiotrophin using multiple mechanisms. Structure 2024; 32:1184-1196.e4. [PMID: 38729161 PMCID: PMC11316656 DOI: 10.1016/j.str.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/21/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024]
Abstract
The integrin Mac-1 (αMβ2, CD11b/CD18, CR3) is an adhesion receptor expressed on macrophages and neutrophils. Mac-1 is also a promiscuous integrin that binds a diverse set of ligands through its αMI-domain. However, the binding mechanism of most ligands remains unclear. We have characterized the interaction of αMI-domain with the cytokine pleiotrophin (PTN), a protein known to bind αMI-domain and induce Mac-1-mediated cell adhesion and migration. Our data show that PTN's N-terminal domain binds a unique site near the N- and C-termini of the αMI-domain using a metal-independent mechanism. However, a stronger interaction is achieved when an acidic amino acid in a zwitterionic motif in PTN's C-terminal domain chelates the divalent cation in the metal ion-dependent adhesion site of active αMI-domain. These results indicate that αMI-domain can bind ligands using multiple mechanisms and that the active αMI-domain has a preference for motifs containing both positively and negatively charged amino acids.
Collapse
Affiliation(s)
- Hoa Nguyen
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85281, USA
| | | | - Tatiana P Ugarova
- School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA
| | - Xu Wang
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85281, USA.
| |
Collapse
|
2
|
Juul-Madsen K, Parbo P, Ismail R, Ovesen PL, Schmidt V, Madsen LS, Thyrsted J, Gierl S, Breum M, Larsen A, Andersen MN, Romero-Ramos M, Holm CK, Andersen GR, Zhao H, Schuck P, Nygaard JV, Sutherland DS, Eskildsen SF, Willnow TE, Brooks DJ, Vorup-Jensen T. Amyloid-β aggregates activate peripheral monocytes in mild cognitive impairment. Nat Commun 2024; 15:1224. [PMID: 38336934 PMCID: PMC10858199 DOI: 10.1038/s41467-024-45627-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
The peripheral immune system is important in neurodegenerative diseases, both in protecting and inflaming the brain, but the underlying mechanisms remain elusive. Alzheimer's Disease is commonly preceded by a prodromal period. Here, we report the presence of large Aβ aggregates in plasma from patients with mild cognitive impairment (n = 38). The aggregates are associated with low level Alzheimer's Disease-like brain pathology as observed by 11C-PiB PET and 18F-FTP PET and lowered CD18-rich monocytes. We characterize complement receptor 4 as a strong binder of amyloids and show Aβ aggregates are preferentially phagocytosed and stimulate lysosomal activity through this receptor in stem cell-derived microglia. KIM127 integrin activation in monocytes promotes size selective phagocytosis of Aβ. Hydrodynamic calculations suggest Aβ aggregates associate with vessel walls of the cortical capillaries. In turn, we hypothesize aggregates may provide an adhesion substrate for recruiting CD18-rich monocytes into the cortex. Our results support a role for complement receptor 4 in regulating amyloid homeostasis.
Collapse
Affiliation(s)
- Kristian Juul-Madsen
- Department of Biomedicine, Aarhus University, The Skou Building, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
- Max-Delbrueck-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Peter Parbo
- Department of Nuclear Medicine, Odense University Hospital, J. B. Winsløws Vej 4, DK-5000, Odense C, Denmark
| | - Rola Ismail
- Department of Nuclear medicine and PET, Vejle Hospital, Beriderbakken 4, DK-7100, Vejle, Denmark
| | - Peter L Ovesen
- Max-Delbrueck-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Vanessa Schmidt
- Max-Delbrueck-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Lasse S Madsen
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11, DK-8200, Aarhus N, Denmark
- Center of Functionally Integrative Neuroscience, Aarhus University and Aarhus University Hospital, Building 1710, Universitetsbyen 3, DK-8200, Aarhus C, Denmark
| | - Jacob Thyrsted
- Department of Biomedicine, Aarhus University, The Skou Building, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Sarah Gierl
- Department of Biomedicine, Aarhus University, The Skou Building, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Mihaela Breum
- Department of Biomedicine, Aarhus University, The Skou Building, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Agnete Larsen
- Department of Biomedicine, Aarhus University, The Skou Building, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Morten N Andersen
- Department of Biomedicine, Aarhus University, The Skou Building, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11, DK-8200, Aarhus N, Denmark
- Department of Hematology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200, Aarhus N, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine, Aarhus University, The Skou Building, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
- NEURODIN AU IDEAS Center, Department of Biomedicine, Aarhus University, The Skou Building, Høegh-Guldbergs Gade 10, DK-8200, Aarhus C, Denmark
| | - Christian K Holm
- Department of Biomedicine, Aarhus University, The Skou Building, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Gregers R Andersen
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, DK-8000, Aarhus C, Denmark
| | - Huaying Zhao
- Laboratory of Dynamics and Macromolecular Assembly, National Institute of Biomedical Imaging and Bioengineering, Building 31, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Peter Schuck
- Laboratory of Dynamics and Macromolecular Assembly, National Institute of Biomedical Imaging and Bioengineering, Building 31, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Jens V Nygaard
- Department of Biological and Chemical Engineering, Aarhus University, Gustav Wieds vej 10 D, DK-8200, Aarhus C, Denmark
| | - Duncan S Sutherland
- Interdisiciplinary Nanoscience Center, Aarhus University, The iNANO House, Gustav Wieds Vej 14, DK-8200, Aarhus C, Denmark
- Center for Cellular Signal Patterns, Aarhus University, The iNANO House, Gustav Wieds Vej 14, DK-8200, Aarhus C, Denmark
| | - Simon F Eskildsen
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11, DK-8200, Aarhus N, Denmark
- Center of Functionally Integrative Neuroscience, Aarhus University and Aarhus University Hospital, Building 1710, Universitetsbyen 3, DK-8200, Aarhus C, Denmark
| | - Thomas E Willnow
- Department of Biomedicine, Aarhus University, The Skou Building, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
- Max-Delbrueck-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - David J Brooks
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200, Aarhus N, Denmark
- Department of Brain Sciences, Imperial College London, Burlington Danes, The Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- Institute of Translational and Clinical Research, University of Newcastle, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Thomas Vorup-Jensen
- Department of Biomedicine, Aarhus University, The Skou Building, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark.
- NEURODIN AU IDEAS Center, Department of Biomedicine, Aarhus University, The Skou Building, Høegh-Guldbergs Gade 10, DK-8200, Aarhus C, Denmark.
- Interdisiciplinary Nanoscience Center, Aarhus University, The iNANO House, Gustav Wieds Vej 14, DK-8200, Aarhus C, Denmark.
| |
Collapse
|
3
|
Nguyen H, Podolnikova NP, Ugarova TP, Wang X. α MI-domain of Integrin Mac-1 Binds the Cytokine Pleiotrophin Using Multiple Mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578455. [PMID: 38352421 PMCID: PMC10862807 DOI: 10.1101/2024.02.01.578455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
The integrin Mac-1 (αMβ2, CD11b/CD18, CR3) is an important adhesion receptor expressed on macrophages and neutrophils. Mac-1 is also the most promiscuous member of the integrin family that binds a diverse set of ligands through its αMI-domain. However, the binding mechanism of most ligands is not clear. We have determined the interaction of αMI-domain with the cytokine pleiotrophin (PTN), a cationic protein known to bind αMI-domain and induce Mac-1-mediated cell adhesion and migration. Our data show that PTN's N-terminal domain binds a unique site near the N- and C-termini of the αMI-domain using a metal-independent mechanism. However, stronger interaction is achieved when an acidic amino acid in a zwitterionic motif in PTN's C-terminal domain chelates the divalent cation in the metal ion-dependent adhesion site of the active αMI-domain. These results indicate that αMI-domain can bind ligands using multiple mechanisms, and suggest that active αMI-domain prefers acidic amino acids in zwitterionic motifs.
Collapse
Affiliation(s)
- Hoa Nguyen
- School of Molecular Sciences, Arizona State University, Tempe, Arizona
| | | | | | - Xu Wang
- School of Molecular Sciences, Arizona State University, Tempe, Arizona
| |
Collapse
|
4
|
Nguyen H, Jing T, Wang X. The Q163C/Q309C mutant of αMI-domain is an active variant suitable for NMR characterization. PLoS One 2023; 18:e0280778. [PMID: 36696377 PMCID: PMC9876370 DOI: 10.1371/journal.pone.0280778] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
Integrin αMβ2 (Mac-1, CD11b/CD18, CR3) is an important adhesion receptor expressed on monocytes. Mac-1 is responsible for mediating cell migration, phagocytosis, degranulation as well as cell-cell fusion. It is also the most promiscuous integrin in terms of ligand specificity with over 100 ligands, most of which use the αMI-domain as their binding site. Despite the importance of αMI-domain in defining ligand interactions of Mac-1, structural studies of αMI-domain's interactions with ligands are lacking. In particular, solution NMR studies of αMI-domain's interaction with ligands have not been possible because the most commonly used active αMI-domain mutants (I316G and ΔK315) are not sufficiently stable and soluble to be used in solution NMR. The goal of this study is to identify an αMI-domain active mutant that's amenable to NMR characterization. By screening known activating mutations of αMI-domain, we determined that the Q163C/Q309C mutant, which converts the αMI-domain into its active form through the formation of an intramolecular disulfide bond, can be produced with a high yield and is more stable than other active mutants. In addition, the Q163C/Q309C mutant has better NMR spectral quality than other active mutants and its affinity for ligands is comparable to other active mutants. Analysis of the Co2+-induced pseudocontact shifts in the Q163C/Q309C mutant showed the structure of the mutant is consistent with the active conformation. Finally, we show that the minor fraction of the Q163C/Q309C mutant without the disulfide bond can be removed through the use of carboxymethyl sepharose chromatography. We think the availability of this mutant for NMR study will significantly enhance structural characterizations of αMI-domain-ligand interactions.
Collapse
Affiliation(s)
- Hoa Nguyen
- School of Molecular Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Tianwei Jing
- Biosensing Instrument Inc., Tempe, Arizona, United States of America
| | - Xu Wang
- School of Molecular Sciences, Arizona State University, Tempe, Arizona, United States of America
| |
Collapse
|
5
|
He Y, Dong L, Yi H, Zhang L, Shi X, Su L, Gan B, Guo R, Wang Y, Luo Q, Li X. Improper preanalytical processes on peripheral blood compromise RNA quality and skew the transcriptional readouts of mRNA and LncRNA. Front Genet 2023; 13:1091685. [PMID: 36685907 PMCID: PMC9845260 DOI: 10.3389/fgene.2022.1091685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Genetic and epigenetic reprogramming caused by disease states in other tissues is always systemically reflected in peripheral blood leukocytes (PBLs). Accurate transcriptional readouts of Messenger RNA (mRNA) and Long non-coding RNA (lncRNA) in peripheral blood leukocytes are fundamental for disease-related study, diagnosis and treatment. However, little is known about the impact of preanalytical variables on RNA quality and downstream messenger RNA and Long non-coding RNA readouts. In this study, we explored the impact of RNA extraction kits and timing of blood placement on peripheral blood leukocyte-derived RNA quality. A novel enhanced evaluation system including RNA yields, purity, RNA integrity number (RIN) values and β-actin copies was employed to more sensitively identify RNA quality differences. The expression levels of informative mRNAs and Long non-coding RNAs in patients with chronic obstructive pulmonary disease (COPD) or triple-negative breast cancer (TNBC) were measured by Quantitative reverse transcription polymerase chain reaction (qRT-PCR) to investigate the impact of RNA quality on transcriptional readouts. Our results showed that the quality of RNA extracted by different kits varies greatly, and commercial kits should be evaluated and managed before batch RNA extraction. In addition, the quality of extracted RNA was highly correlated with the timing of blood placement, and the copy number of β-actin was significantly decreased after leaving blood at RT over 12 h. More importantly, compromised RNA leads to skewed transcriptional readouts of informative mRNAs and Long non-coding RNAs in patients with chronic obstructive pulmonary disease or triple-negative breast cancer. These findings have significant implications for peripheral blood leukocyte-derived RNA quality management and suggest that quality control is necessary prior to the analysis of patient messenger RNA and Long non-coding RNA expression.
Collapse
Affiliation(s)
- Yinli He
- BioBank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Lele Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Hongyang Yi
- National Clinical Research Centre for Infectious Diseases, The Third People’s Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Linpei Zhang
- BioBank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xue Shi
- BioBank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Lin Su
- BioBank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Baoyu Gan
- BioBank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Ruirui Guo
- BioBank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yawen Wang
- BioBank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China,*Correspondence: Xiaojiao Li, ; Qinying Luo, ; Yawen Wang,
| | - Qinying Luo
- BioBank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China,*Correspondence: Xiaojiao Li, ; Qinying Luo, ; Yawen Wang,
| | - Xiaojiao Li
- BioBank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China,*Correspondence: Xiaojiao Li, ; Qinying Luo, ; Yawen Wang,
| |
Collapse
|
6
|
Casteel JL, Keever KR, Ardell CL, Williams DL, Gao D, Podrez EA, Byzova TV, Yakubenko VP. Modification of Extracellular Matrix by the Product of DHA Oxidation Switches Macrophage Adhesion Patterns and Promotes Retention of Macrophages During Chronic Inflammation. Front Immunol 2022; 13:867082. [PMID: 35720381 PMCID: PMC9204313 DOI: 10.3389/fimmu.2022.867082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Oxidation of polyunsaturated fatty acids contributes to different aspects of the inflammatory response due to the variety of products generated. Specifically, the oxidation of DHA produces the end-product, carboxyethylpyrrole (CEP), which forms a covalent adduct with proteins via an ϵ-amino group of lysines. Previously, we found that CEP formation is dramatically increased in inflamed tissue and CEP-modified albumin and fibrinogen became ligands for αDβ2 (CD11d/CD18) and αMβ2 (CD11b/CD18) integrins. In this study, we evaluated the effect of extracellular matrix (ECM) modification with CEP on the adhesive properties of M1-polarized macrophages, particularly during chronic inflammation. Using digested atherosclerotic lesions and in vitro oxidation assays, we demonstrated the ability of ECM proteins to form adducts with CEP, particularly, DHA oxidation leads to the formation of CEP adducts with collagen IV and laminin, but not with collagen I. Using integrin αDβ2-transfected HEK293 cells, WT and α D - / - mouse M1-polarized macrophages, we revealed that CEP-modified proteins support stronger cell adhesion and spreading when compared with natural ECM ligands such as collagen IV, laminin, and fibrinogen. Integrin αDβ2 is critical for M1 macrophage adhesion to CEP. Based on biolayer interferometry results, the isolated αD I-domain demonstrates markedly higher binding affinity to CEP compared to the "natural" αDβ2 ligand fibrinogen. Finally, the presence of CEP-modified proteins in a 3D fibrin matrix significantly increased M1 macrophage retention. Therefore, CEP modification converts ECM proteins to αDβ2-recognition ligands by changing a positively charged lysine to negatively charged CEP, which increases M1 macrophage adhesion to ECM and promotes macrophage retention during detrimental inflammation, autoimmunity, and chronic inflammation.
Collapse
Affiliation(s)
- Jared L. Casteel
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Kasey R. Keever
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Christopher L. Ardell
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - David L. Williams
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Detao Gao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Eugene A. Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Tatiana V. Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Valentin P. Yakubenko
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
7
|
The crystal structure of iC3b-CR3 αI reveals a modular recognition of the main opsonin iC3b by the CR3 integrin receptor. Nat Commun 2022; 13:1955. [PMID: 35413960 PMCID: PMC9005620 DOI: 10.1038/s41467-022-29580-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/15/2022] [Indexed: 12/27/2022] Open
Abstract
Complement activation on cell surfaces leads to the massive deposition of C3b, iC3b, and C3dg, the main complement opsonins. Recognition of iC3b by complement receptor type 3 (CR3) fosters pathogen opsonophagocytosis by macrophages and the stimulation of adaptive immunity by complement-opsonized antigens. Here, we present the crystallographic structure of the complex between human iC3b and the von Willebrand A inserted domain of the α chain of CR3 (αI). The crystal contains two composite interfaces for CR3 αI, encompassing distinct sets of contiguous macroglobulin (MG) domains on the C3c moiety, MG1-MG2 and MG6-MG7 domains. These composite binding sites define two iC3b-CR3 αI complexes characterized by specific rearrangements of the two semi-independent modules, C3c moiety and TED domain. Furthermore, we show the structure of iC3b in a physiologically-relevant extended conformation. Based on previously available data and novel insights reported herein, we propose an integrative model that reconciles conflicting facts about iC3b structure and function and explains the molecular basis for iC3b selective recognition by CR3 on opsonized surfaces. Complement activation on foreign cell surfaces leads to the generation of complement opsonins, which activate complement receptor type 3 (CR3) and pathogen clearance by macrophages. Here, the authors reveal structural basis of the interaction between human opsonin iC3b and the von Willebrand A inserted domain of the α chain of CR3.
Collapse
|
8
|
Liu D, Duan L, Cyster JG. Chemo- and mechanosensing by dendritic cells facilitate antigen surveillance in the spleen. Immunol Rev 2022; 306:25-42. [PMID: 35147233 PMCID: PMC8852366 DOI: 10.1111/imr.13055] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/05/2021] [Indexed: 12/30/2022]
Abstract
Spleen dendritic cells (DC) are critical for initiation of adaptive immune responses against blood-borne invaders. Key to DC function is their positioning at sites of pathogen entry, and their abilities to selectively capture foreign antigens and promptly engage T cells. Focusing on conventional DC2 (cDC2), we discuss the contribution of chemoattractant receptors (EBI2 or GPR183, S1PR1, and CCR7) and integrins to cDC2 positioning and function. We give particular attention to a newly identified role in cDC2 for adhesion G-protein coupled receptor E5 (Adgre5 or CD97) and its ligand CD55, detailing how this mechanosensing system contributes to splenic cDC2 positioning and homeostasis. Additional roles of CD97 in the immune system are reviewed. The ability of cDC2 to be activated by circulating missing self-CD47 cells and to integrate multiple red blood cell (RBC)-derived inputs is discussed. Finally, we describe the process of activated cDC2 migration to engage and prime helper T cells. Throughout the review, we consider the insights into cDC function in the spleen that have emerged from imaging studies.
Collapse
Affiliation(s)
- Dan Liu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| | - Lihui Duan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| |
Collapse
|
9
|
On the Use of Surface Plasmon Resonance Biosensing to Understand IgG-FcγR Interactions. Int J Mol Sci 2021; 22:ijms22126616. [PMID: 34205578 PMCID: PMC8235063 DOI: 10.3390/ijms22126616] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 01/01/2023] Open
Abstract
Surface plasmon resonance (SPR)-based optical biosensors offer real-time and label-free analysis of protein interactions, which has extensively contributed to the discovery and development of therapeutic monoclonal antibodies (mAbs). As the biopharmaceutical market for these biologics and their biosimilars is rapidly growing, the role of SPR biosensors in drug discovery and quality assessment is becoming increasingly prominent. One of the critical quality attributes of mAbs is the N-glycosylation of their Fc region. Other than providing stability to the antibody, the Fc N-glycosylation influences immunoglobulin G (IgG) interactions with the Fcγ receptors (FcγRs), modulating the immune response. Over the past two decades, several studies have relied on SPR-based assays to characterize the influence of N-glycosylation upon the IgG-FcγR interactions. While these studies have unveiled key information, many conclusions are still debated in the literature. These discrepancies can be, in part, attributed to the design of the reported SPR-based assays as well as the methodology applied to SPR data analysis. In fact, the SPR biosensor best practices have evolved over the years, and several biases have been pointed out in the development of experimental SPR protocols. In parallel, newly developed algorithms and data analysis methods now allow taking into consideration complex biomolecular kinetics. In this review, we detail the use of different SPR biosensing approaches for characterizing the IgG-FcγR interactions, highlighting their merit and inherent experimental complexity. Furthermore, we review the latest SPR-derived conclusions on the influence of the N-glycosylation upon the IgG-FcγR interactions and underline the differences and similarities across the literature. Finally, we explore new avenues taking advantage of novel computational analysis of SPR results as well as the latest strategies to control the glycoprofile of mAbs during production, which could lead to a better understanding and modelling of the IgG-FcγRs interactions.
Collapse
|
10
|
CD11c regulates hematopoietic stem and progenitor cells under stress. Blood Adv 2021; 4:6086-6097. [PMID: 33351105 DOI: 10.1182/bloodadvances.2020002504] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/09/2020] [Indexed: 12/28/2022] Open
Abstract
β2 integrins are well-known leukocyte adhesion molecules consisting of 4 members: CD11a-d. Their known biological functions range widely from leukocyte recruitment, phagocytosis, to immunological synapse formation, but the studies have been primarily focused on CD11a and CD11b. CD11c is 1 of the 4 members and is extremely homologous to CD11b. It has been well known as a dendritic cell marker, but the characterization of its function has been limited. We found that CD11c was expressed on the short-term hematopoietic stem cells and multipotent progenitor cells. The lack of CD11c did not affect the number of hematopoietic stem and progenitor cells (HSPCs) in healthy CD11c knockout mice. Different from other β2 integrin members, however, CD11c deficiency was associated with increased apoptosis and significant loss of HSPCs in sepsis and bone marrow transplantation. Although integrins are generally known for their overlapping and redundant roles, we showed that CD11c had a distinct role of regulating the expansion of HSPCs under stress. This study shows that CD11c, a well-known dendritic cell marker, is expressed on HSPCs and serves as their functional regulator. CD11c deficiency leads to the loss of HSPCs via apoptosis in sepsis and bone marrow transplantation.
Collapse
|
11
|
Wang F, Ullah A, Fan X, Xu Z, Zong R, Wang X, Chen G. Delivery of nanoparticle antigens to antigen-presenting cells: from extracellular specific targeting to intracellular responsive presentation. J Control Release 2021; 333:107-128. [PMID: 33774119 DOI: 10.1016/j.jconrel.2021.03.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/05/2023]
Abstract
An appropriate delivery system can improve the immune effects of antigens against various infections or tumors. Antigen-presenting cells (APCs) are specialized to capture and process antigens in vivo, which link the innate and adaptive immune responses. Functionalization of vaccine delivery systems with targeting moieties to APCs is a promising strategy for provoking potent immune responses. Additionally, the internalization and intracellular distribution of antigens are closely related to the initiation of downstream immune responses. With a deeper understanding of the intracellular microenvironment and the mechanisms of antigen presentation, vehicles designed to respond to endogenous and external stimuli can modulate antigen processing and presentation pathways, which are critical to the types of immune response. Here, an overview of extracellular targeting delivery of antigens to APCs and intracellular stimulus-responsiveness strategies is provided, which might be helpful for the rational design of vaccine delivery systems.
Collapse
Affiliation(s)
- Fei Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Aftab Ullah
- Shantou University Medical College, Shantou 515041, China
| | - Xuelian Fan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Zhou Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Rongling Zong
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Xuewen Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Gang Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
12
|
Choi J, Buyannemekh D, Nham SU. Moieties of Complement iC3b Recognized by the I-domain of Integrin αXβ2. Mol Cells 2020; 43:1023-1034. [PMID: 33372665 PMCID: PMC7772510 DOI: 10.14348/molcells.2020.0197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/17/2020] [Indexed: 11/27/2022] Open
Abstract
Complement fragment iC3b serves as a major opsonin for facilitating phagocytosis via its interaction with complement receptors CR3 and CR4, also known by their leukocyte integrin family names, αMβ2 and αXβ2, respectively. Although there is general agreement that iC3b binds to the αM and αX I-domains of the respective β2-integrins, much less is known regarding the regions of iC3b contributing to the αX I-domain binding. In this study, using recombinant αX I-domain, as well as recombinant fragments of iC3b as candidate binding partners, we have identified two distinct binding moieties of iC3b for the αX I-domain. They are the C3 convertase-generated N-terminal segment of the C3b α'- chain (α'NT) and the factor I cleavage-generated N-terminal segment in the CUBf region of α-chain. Additionally, we have found that the CUBf segment is a novel binding moiety of iC3b for the αM I-domain. The CUBf segment shows about a 2-fold higher binding activity than the α'NT for αX I-domain. We also have shown the involvement of crucial acidic residues on the iC3b side of the interface and basic residues on the I-domain side.
Collapse
Affiliation(s)
- Jeongsuk Choi
- Department of Biology, Kangwon National University, Chuncheon 2434, Korea
| | | | - Sang-Uk Nham
- Division of Science Education, Kangwon National University, Chuncheon 4341, Korea
| |
Collapse
|
13
|
Combes F, Meyer E, Sanders NN. Immune cells as tumor drug delivery vehicles. J Control Release 2020; 327:70-87. [PMID: 32735878 DOI: 10.1016/j.jconrel.2020.07.043] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/21/2022]
Abstract
This review article describes the use of immune cells as potential candidates to deliver anti-cancer drugs deep within the tumor microenvironment. First, the rationale of using drug carriers to target tumors and potentially decrease drug-related side effects is discussed. We further explain some of the current limitations when using nanoparticles for this purpose. Next, a comprehensive step-by-step description of the migration cascade of immune cells is provided as well as arguments on why immune cells can be used to address some of the limitations associated with nanoparticle-mediated drug delivery. We then describe the benefits and drawbacks of using red blood cells, platelets, granulocytes, monocytes, macrophages, myeloid-derived suppressor cells, T cells and NK cells for tumor-targeted drug delivery. An additional section discusses the versatility of nanoparticles to load anti-cancer drugs into immune cells. Lastly, we propose increasing the circulatory half-life and development of conditional release strategies as the two main future pillars to improve the efficacy of immune cell-mediated drug delivery to tumors.
Collapse
Affiliation(s)
- Francis Combes
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Evelyne Meyer
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium.
| |
Collapse
|
14
|
Klug D, Goellner S, Kehrer J, Sattler J, Strauss L, Singer M, Lu C, Springer TA, Frischknecht F. Evolutionarily distant I domains can functionally replace the essential ligand-binding domain of Plasmodium TRAP. eLife 2020; 9:57572. [PMID: 32648541 PMCID: PMC7351488 DOI: 10.7554/elife.57572] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/25/2020] [Indexed: 02/02/2023] Open
Abstract
Inserted (I) domains function as ligand-binding domains in adhesins that support cell adhesion and migration in many eukaryotic phyla. These adhesins include integrin αβ heterodimers in metazoans and single subunit transmembrane proteins in apicomplexans such as TRAP in Plasmodium and MIC2 in Toxoplasma. Here we show that the I domain of TRAP is essential for sporozoite gliding motility, mosquito salivary gland invasion and mouse infection. Its replacement with the I domain from Toxoplasma MIC2 fully restores tissue invasion and parasite transmission, while replacement with the aX I domain from human integrins still partially restores liver infection. Mutations around the ligand binding site allowed salivary gland invasion but led to inefficient transmission to the rodent host. These results suggest that apicomplexan parasites appropriated polyspecific I domains in part for their ability to engage with multiple ligands and to provide traction for emigration into diverse organs in distant phyla. Malaria is an infectious disease caused by single-celled parasites known as Plasmodium. Humans and other animals with backbones – such as birds, reptiles and rodents – can become hosts for these parasites if an infected female mosquito feeds on their blood. Likewise, healthy mosquitoes can in turn become infected with Plasmodium if they feed on the blood of an infected animal. To complete their life cycle, Plasmodium parasites within a mosquito must become spore-like cells called sporozoites. These sporozoites are highly mobile and can get into the mosquitoes’ salivary glands, meaning they can be passed on to a new host when the insect feeds. During a mosquito bite the sporozoites are spat into the skin of the potential host, where they then need to migrate rapidly to enter the bloodstream. Once in the blood, the sporozoites can then get into liver cells and begin a new infection. One protein called TRAP, which is found on the surface of the sporozoites, is important for their migration and the infection of the salivary glands or liver. Yet it was not known how this happens at the level of the individual proteins involved. Klug et al. have now tested how a part of the TRAP protein, called the I domain, contributes to the infection process. In the experiments, the I domain of TRAP was deleted which showed that the sporozoites need this domain to be able to move around and get into the host tissues. Without the I domain the sporozoites were stuck and could not successfully infect either the mosquitoes, the livers of mice, or human liver cells grown in the laboratory. Klug et al. then replaced the Plasmodium I domain of TRAP with the I domain from a distantly related parasite called Toxoplasma gondii, which causes a condition known as toxoplasmosis. The I domain from Toxoplasma allowed the Plasmodium parasites to infect the host tissues again. This observation was unexpected because Toxoplasma and Plasmodium parasites have evolved separately over the last 800 million years and Toxoplasma does not infect insects. These findings suggest that the I domain of TRAP evolved to bind several other proteins in different tissues and hosts. Future studies will investigate which other parasite proteins TRAP works with to guide sporozoites to the salivary glands or liver. Knowledge of how these proteins act together may lead to new approaches for treating or preventing malaria. For example, some treatments could stop sporozoites from entering liver cells.
Collapse
Affiliation(s)
- Dennis Klug
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany.,Université de Strasbourg, CNRS UPR9022, INSERM U963, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Sarah Goellner
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany.,Department of Molecular Virology, Heidelberg University Medical School, Heidelberg, Germany
| | - Jessica Kehrer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Julia Sattler
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Léanne Strauss
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Mirko Singer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany.,Experimental Parasitology, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, München, Germany
| | - Chafen Lu
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
| | - Timothy A Springer
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| |
Collapse
|
15
|
Juul-Madsen K, Qvist P, Bendtsen KL, Langkilde AE, Vestergaard B, Howard KA, Dehesa-Etxebeste M, Paludan SR, Andersen GR, Jensen PH, Otzen DE, Romero-Ramos M, Vorup-Jensen T. Size-Selective Phagocytic Clearance of Fibrillar α-Synuclein through Conformational Activation of Complement Receptor 4. THE JOURNAL OF IMMUNOLOGY 2020; 204:1345-1361. [PMID: 31969389 DOI: 10.4049/jimmunol.1900494] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 12/18/2019] [Indexed: 12/19/2022]
Abstract
Aggregation of α-synuclein (αSN) is an important histological feature of Parkinson disease. Recent studies showed that the release of misfolded αSN from human and rodent neurons is relevant to the progression and spread of αSN pathology. Little is known, however, about the mechanisms responsible for clearance of extracellular αSN. This study found that human complement receptor (CR) 4 selectively bound fibrillar αSN, but not monomeric species. αSN is an abundant protein in the CNS, which potentially could overwhelm clearance of cytotoxic αSN species. The selectivity of CR4 toward binding fibrillar αSN consequently adds an important αSN receptor function for maintenance of brain homeostasis. Based on the recently solved structures of αSN fibrils and the known ligand preference of CR4, we hypothesize that the parallel monomer stacking in fibrillar αSN creates a known danger-associated molecular pattern of stretches of anionic side chains strongly bound by CR4. Conformational change in the receptor regulated tightly clearance of fibrillar αSN by human monocytes. The induced change coupled concomitantly with phagolysosome formation. Data mining of the brain transcriptome in Parkinson disease patients supported CR4 as an active αSN clearance mechanism in this disease. Our results associate an important part of the innate immune system, namely complement receptors, with the central molecular mechanisms of CNS protein aggregation in neurodegenerative disorders.
Collapse
Affiliation(s)
- Kristian Juul-Madsen
- Biophysical Immunology Laboratory, Aarhus University, DK-8000 Aarhus C, Denmark.,Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Per Qvist
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark.,iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus University, DK-8000 Aarhus C, Denmark.,iSEQ, Centre for Integrative Sequencing, Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Kirstine L Bendtsen
- Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen Ø, Denmark
| | - Annette E Langkilde
- Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen Ø, Denmark
| | - Bente Vestergaard
- Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen Ø, Denmark
| | - Kenneth A Howard
- Interdisciplinary Nanoscience Center, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Martxel Dehesa-Etxebeste
- Neuroscience Area, Biodonostia Research Institute, 20014 Donostia, San Sebastian, Spain.,CIBERNED, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Søren R Paludan
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Gregers Rom Andersen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Poul Henning Jensen
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark.,DANDRITE-Danish Research Institute of Translational Neuroscience, Aarhus University, DK-8000 Aarhus C, Denmark; and
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Center, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark.,DANDRITE-Danish Research Institute of Translational Neuroscience, Aarhus University, DK-8000 Aarhus C, Denmark; and.,NEURODIN AU IDEAS Center, Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Thomas Vorup-Jensen
- Biophysical Immunology Laboratory, Aarhus University, DK-8000 Aarhus C, Denmark; .,Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark.,Interdisciplinary Nanoscience Center, Aarhus University, DK-8000 Aarhus C, Denmark.,NEURODIN AU IDEAS Center, Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
16
|
Vorup-Jensen T, Jensen RK. Structural Immunology of Complement Receptors 3 and 4. Front Immunol 2018; 9:2716. [PMID: 30534123 PMCID: PMC6275225 DOI: 10.3389/fimmu.2018.02716] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 11/05/2018] [Indexed: 01/10/2023] Open
Abstract
Complement receptors (CR) 3 and 4 belong to the family of beta-2 (CD18) integrins. CR3 and CR4 are often co-expressed in the myeloid subsets of leukocytes, but they are also found in NK cells and activated T and B lymphocytes. The heterodimeric ectodomain undergoes considerable conformational change in order to switch the receptor from a structurally bent, ligand-binding in-active state into an extended, ligand-binding active state. CR3 binds the C3d fragment of C3 in a way permitting CR2 also to bind concomitantly. This enables a hand-over of complement-opsonized antigens from the cell surface of CR3-expressing macrophages to the CR2-expressing B lymphocytes, in consequence acting as an antigen presentation mechanism. As a more enigmatic part of their functions, both CR3 and CR4 bind several structurally unrelated proteins, engineered peptides, and glycosaminoglycans. No consensus motif in the proteinaceous ligands has been established. Yet, the experimental evidence clearly suggest that the ligands are primarily, if not entirely, recognized by a single site within the receptors, namely the metal-ion dependent adhesion site (MIDAS). Comparison of some recent identified ligands points to CR3 as inclined to bind positively charged species, while CR4, by contrast, binds strongly negative-charged species, in both cases with the critical involvement of deprotonated, acidic groups as ligands for the Mg2+ ion in the MIDAS. These properties place CR3 and CR4 firmly within the realm of modern molecular medicine in several ways. The expression of CR3 and CR4 in NK cells was recently demonstrated to enable complement-dependent cell cytotoxicity toward antibody-coated cancer cells as part of biological therapy, constituting a significant part of the efficacy of such treatment. With the flexible principles of ligand recognition, it is also possible to propose a response of CR3 and CR4 to existing medicines thereby opening a possibility of drug repurposing to influence the function of these receptors. Here, from advances in the structural and cellular immunology of CR3 and CR4, we review insights on their biochemistry and functions in the immune system.
Collapse
Affiliation(s)
- Thomas Vorup-Jensen
- Biophysical Immunology Laboratory, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| | - Rasmus Kjeldsen Jensen
- Department of Molecular Biology and Genetics-Structural Biology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
17
|
Heng Z, Ruan L, Gan R. Three Methods to Purify Leukocytes and RNA Quality Assessment. Biopreserv Biobank 2018; 16:434-438. [PMID: 30379576 DOI: 10.1089/bio.2018.0058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Leukocytes function as central effectors in innate immunity (such as phagocytosis) as well as adaptive immunity (e.g., antigen-dependent T cell activation), and serve as an important resource in the fields of translational medicine, precision medicine, and cell therapy. Isolation of leukocytes from whole blood is necessary for high-quality RNA and downstream research. This process is susceptible to the variability of many factors, such as blood collection, isolation reagents, and extraction methods. In this study, three methods were applied for leukocytes separation, followed by RNA extraction and quality testing to evaluate the methods. Results showed that leukocytes were purified using lymphocyte separation medium (LSM), optimized LSM method, or red blood cell lysis buffer (RBC lysis), and RNA quality met the basic requirements for downstream studies. Although considering the simplicity of the procedure and RNA quality from donated samples, the RBC lysis method should be recommended to biobanks for further research.
Collapse
Affiliation(s)
- Zhang Heng
- Shanghai Clinical Research Center, Shanghai, China.,Shanghai Engineering Research Center of Biobank, Shanghai, China
| | - Liangliang Ruan
- Shanghai Clinical Research Center, Shanghai, China.,Shanghai Engineering Research Center of Biobank, Shanghai, China
| | - Rongxing Gan
- Shanghai Clinical Research Center, Shanghai, China.,Shanghai Engineering Research Center of Biobank, Shanghai, China
| |
Collapse
|
18
|
Yakubenko VP, Cui K, Ardell CL, Brown KE, West XZ, Gao D, Stefl S, Salomon RG, Podrez EA, Byzova TV. Oxidative modifications of extracellular matrix promote the second wave of inflammation via β 2 integrins. Blood 2018; 132:78-88. [PMID: 29724896 PMCID: PMC6034644 DOI: 10.1182/blood-2017-10-810176] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 04/19/2018] [Indexed: 12/13/2022] Open
Abstract
Early stages of inflammation are characterized by extensive oxidative insult by recruited and activated neutrophils. Secretion of peroxidases, including the main enzyme, myeloperoxidase, leads to the generation of reactive oxygen species. We show that this oxidative insult leads to polyunsaturated fatty acid (eg, docosahexaenoate), oxidation, and accumulation of its product 2-(ω-carboxyethyl)pyrrole (CEP), which, in turn, is capable of protein modifications. In vivo CEP is generated predominantly at the inflammatory sites in macrophage-rich areas. During thioglycollate-induced inflammation, neutralization of CEP adducts dramatically reduced macrophage accumulation in the inflamed peritoneal cavity while exhibiting no effect on the early recruitment of neutrophils, suggesting a role in the second wave of inflammation. CEP modifications were abundantly deposited along the path of neutrophils migrating through the 3-dimensional fibrin matrix in vitro. Neutrophil-mediated CEP formation was markedly inhibited by the myeloperoxidase inhibitor, 4-ABH, and significantly reduced in myeloperoxidase-deficient mice. On macrophages, CEP adducts were recognized by cell adhesion receptors, integrin αMβ2 and αDβ2 Macrophage migration through CEP-fibrin gel was dramatically augmented when compared with fibrin alone, and was reduced by β2-integrin deficiency. Thus, neutrophil-mediated oxidation of abundant polyunsaturated fatty acids leads to the transformation of existing proteins into stronger adhesive ligands for αMβ2- and αDβ2-dependent macrophage migration. The presence of a carboxyl group rather than a pyrrole moiety on these adducts, resembling characteristics of bacterial and/or immobilized ligands, is critical for recognition by macrophages. Therefore, specific oxidation-dependent modification of extracellular matrix, aided by neutrophils, promotes subsequent αMβ2- and αDβ2-mediated migration/retention of macrophages during inflammation.
Collapse
Affiliation(s)
- Valentin P Yakubenko
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH; and the
| | - Kui Cui
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN
| | - Christopher L Ardell
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN
| | - Kathleen E Brown
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH; and the
| | - Xiaoxia Z West
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH; and the
| | - Detao Gao
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH; and the
| | - Samantha Stefl
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH; and the
| | - Robert G Salomon
- Department of Chemistry, Case Western Reserve University, Cleveland, OH
| | - Eugene A Podrez
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH; and the
| | - Tatiana V Byzova
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH; and the
| |
Collapse
|
19
|
Thinn AMM, Wang Z, Zhu J. The membrane-distal regions of integrin α cytoplasmic domains contribute differently to integrin inside-out activation. Sci Rep 2018; 8:5067. [PMID: 29568062 PMCID: PMC5864728 DOI: 10.1038/s41598-018-23444-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/13/2018] [Indexed: 12/20/2022] Open
Abstract
Functioning as signal receivers and transmitters, the integrin α/β cytoplasmic tails (CT) are pivotal in integrin activation and signaling. 18 α integrin subunits share a conserved membrane-proximal region but have a highly diverse membrane-distal (MD) region at their CTs. Recent studies demonstrated that the presence of α CTMD region is essential for talin-induced integrin inside-out activation. However, it remains unknown whether the non-conserved α CTMD regions differently regulate the inside-out activation of integrin. Using αIIbβ3, αLβ2, and α5β1 as model integrins and by replacing their α CTMD regions with those of α subunits that pair with β3, β2, and β1 subunits, we analyzed the function of CTMD regions of 17 α subunits in talin-mediated integrin activation. We found that the α CTMD regions play two roles on integrin, which are activation-supportive and activation-regulatory. The regulatory but not the supportive function depends on the sequence identity of α CTMD region. A membrane-proximal tyrosine residue present in the CTMD regions of a subset of α integrins was identified to negatively regulate integrin inside-out activation. Our study provides a useful resource for investigating the function of α integrin CTMD regions.
Collapse
Affiliation(s)
- Aye Myat Myat Thinn
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Zhengli Wang
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, 53226, USA
| | - Jieqing Zhu
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, 53226, USA.
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
20
|
Abstract
Leukocytes can completely reorganize their cytoskeletal architecture within minutes. This structural plasticity, which facilitates their migration and communicative function, also enables them to exert a substantial amount of mechanical force against the extracellular matrix and the surfaces of interacting cells. In recent years, it has become increasingly clear that these forces have crucial roles in immune cell activation and subsequent effector responses. Here, I review our current understanding of how mechanical force regulates cell-surface receptor activation, cell migration, intracellular signalling and intercellular communication, highlighting the biological ramifications of these effects in various immune cell types.
Collapse
Affiliation(s)
- Morgan Huse
- Immunology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| |
Collapse
|
21
|
Abstract
Myeloid cells make extensive use of the complement system in the context of recruitment, phagocytosis, and other effector functions. There are several types of complement receptors on myeloid cells, including G protein-coupled receptors for localizing the source of complement activation, and three sets of type I transmembrane proteins that link complement to phagocytosis: complement receptor 1, having an extracellular domain with tandem complement regulatory repeats; complement receptors 3 and 4, which are integrin family receptors comprising heterodimers of type I transmembrane subunits; and VSIG4, a member of the Ig superfamily. This review will focus on the role of the different classes of complement receptors and how their activities are integrated in the setting of immune tolerance and inflammatory responses.
Collapse
|
22
|
Buyannemekh D, Nham SU. Characterization of αX I-Domain Binding to Receptors for Advanced Glycation End Products (RAGE). Mol Cells 2017; 40:355-362. [PMID: 28535664 PMCID: PMC5463044 DOI: 10.14348/molcells.2017.0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/13/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023] Open
Abstract
The β2 integrins are cell surface transmembrane proteins regulating leukocyte functions, such as adhesion and migration. Two members of β2 integrin, αMβ2 and αXβ2, share the leukocyte distribution profile and integrin αXβ2 is involved in antigen presentation in dendritic cells and transendothelial migration of monocytes and macrophages to atherosclerotic lesions. Receptor for advanced glycation end products (RAGE), a member of cell adhesion molecules, plays an important role in chronic inflammation and atherosclerosis. Although RAGE and αXβ2 play an important role in inflammatory response and the pathogenesis of atherosclerosis, the nature of their interaction and structure involved in the binding remain poorly defined. In this study, using I-domain as a ligand binding motif of αXβ2, we characterize the binding nature and the interacting moieties of αX I-domain and RAGE. Their binding requires divalent cations (Mg2+ and Mn2+) and shows an affinity on the sub-micro molar level: the dissociation constant of αX I-domains binding to RAGE being 0.49 μM. Furthermore, the αX I-domains recognize the V-domain, but not the C1 and C2-domains of RAGE. The acidic amino acid substitutions on the ligand binding site of αX I-domain significantly reduce the I-domain binding activity to soluble RAGE and the alanine substitutions of basic amino acids on the flat surface of the V-domain prevent the V-domain binding to αX I-domain. In conclusion, the main mechanism of αX I-domain binding to RAGE is a charge interaction, in which the acidic moieties of αX I-domains, including E244, and D249, recognize the basic residues on the RAGE V-domain encompassing K39, K43, K44, R104, and K107.
Collapse
Affiliation(s)
- Dolgorsuren Buyannemekh
- Divisions of Science Education and Biology, Kangwon National University, Chuncheon, Chuncheon 24341,
Korea
| | - Sang-Uk Nham
- Divisions of Science Education and Biology, Kangwon National University, Chuncheon, Chuncheon 24341,
Korea
| |
Collapse
|
23
|
Pakula MM, Maier TJ, Vorup-Jensen T. Insight on the impacts of free amino acids and their metabolites on the immune system from a perspective of inborn errors of amino acid metabolism. Expert Opin Ther Targets 2017; 21:611-626. [PMID: 28441889 DOI: 10.1080/14728222.2017.1323879] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Amino acids (AAs) support a broad range of functions in living organisms, including several that affect the immune system. The functions of the immune system are affected when free AAs are depleted or in excess because of external factors, such as starvation, or because of genetic factors, such as inborn errors of metabolism. Areas covered: In this review, we discuss the current insights into how free AAs affect immune responses. When possible, we make comparisons to known disease states resulting from inborn errors of metabolism, in which changed levels of AAs or AA metabolites provide insight into the impact of AAs on the human immune system in vivo. We also explore the literature describing how changes in AA levels might provide pharmaceutical targets for safe immunomodulatory treatment. Expert opinion: The impact of free AAs on the immune system is a neglected topic in most immunology textbooks. That neglect is undeserved, because free AAs have both direct and indirect effects on the immune system. Consistent choices of pre-clinical models and better strategies for creating formulations are required to gain clinical impact.
Collapse
Affiliation(s)
| | - Thorsten J Maier
- a Department of Biomedicine , Aarhus University , Aarhus , Denmark
| | - Thomas Vorup-Jensen
- a Department of Biomedicine , Aarhus University , Aarhus , Denmark.,b Center for Neurodegenerative Inflammation Prevention (NEURODIN) , Aarhus University , Aarhus , Denmark.,c Interdisciplinary Nanoscience Center , Aarhus University , Aarhus , Denmark.,d The Lundbeck Foundation Nanomedicine Center for Individualized Management of Tissue Damage and Regeneration (LUNA) , Aarhus University , Aarhus , Denmark.,e MEMBRANES Research center , Aarhus University , Aarhus , Denmark
| |
Collapse
|
24
|
Distinct recognition of complement iC3b by integrins α Xβ 2 and α Mβ 2. Proc Natl Acad Sci U S A 2017; 114:3403-3408. [PMID: 28292891 DOI: 10.1073/pnas.1620881114] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Recognition by the leukocyte integrins αXβ2 and αMβ2 of complement iC3b-opsonized targets is essential for effector functions including phagocytosis. The integrin-binding sites on iC3b remain incompletely characterized. Here, we describe negative-stain electron microscopy and biochemical studies of αXβ2 and αMβ2 in complex with iC3b. Despite high homology, the two integrins bind iC3b at multiple distinct sites. αXβ2 uses the αX αI domain to bind iC3b on its C3c moiety at one of two sites: a major site at the interface between macroglobulin (MG) 3 and MG4 domains, and a less frequently used site near the C345C domain. In contrast, αMβ2 uses its αI domain to bind iC3b at the thioester domain and simultaneously interacts through a region near the αM β-propeller and β2 βI domain with a region of the C3c moiety near the C345C domain. Remarkably, there is no overlap between the primary binding site of αXβ2 and the binding site of αMβ2 on iC3b. Distinctive binding sites on iC3b by integrins αXβ2 and αMβ2 may be biologically beneficial for leukocytes to more efficiently capture opsonized pathogens and to avoid subversion by pathogen factors.
Collapse
|
25
|
Abstract
Mac-1 (CD11b/CD18) is a β2 integrin classically regarded as a pro-inflammatory molecule because of its ability to promote phagocyte cytotoxic functions and enhance the function of several effector molecules such as FcγR, uPAR, and CD14. Nevertheless, recent reports have revealed that Mac-1 also plays significant immunoregulatory roles, and genetic variants in ITGAM, the gene that encodes CD11b, confer risk for the autoimmune disease systemic lupus erythematosus (SLE). This has renewed interest in the physiological roles of this integrin and raised new questions on how its seemingly opposing biological functions may be regulated. Here, we provide an overview of the CD18 integrins and how their activation may be regulated as this may shed light on how the opposing roles of Mac-1 may be elicited. We then discuss studies that exemplify Mac-1's pro-inflammatory versus regulatory roles particularly in the context of IgG immune complex-mediated inflammation. This includes a detailed examination of molecular mechanisms that could explain the risk-conferring effect of rs1143679, a single nucleotide non-synonymous Mac-1 polymorphism associated with SLE.
Collapse
Affiliation(s)
- Florencia Rosetti
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Tanya N Mayadas
- Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Sándor N, Lukácsi S, Ungai-Salánki R, Orgován N, Szabó B, Horváth R, Erdei A, Bajtay Z. CD11c/CD18 Dominates Adhesion of Human Monocytes, Macrophages and Dendritic Cells over CD11b/CD18. PLoS One 2016; 11:e0163120. [PMID: 27658051 PMCID: PMC5033469 DOI: 10.1371/journal.pone.0163120] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/03/2016] [Indexed: 12/13/2022] Open
Abstract
Complement receptors CR3 (CD11b/CD18) and CR4 (CD11c/CD18) belong to the family of beta2 integrins and are expressed mainly by myeloid cell types in humans. Previously, we proved that CR3 rather than CR4 plays a key role in phagocytosis. Here we analysed how CD11b and CD11c participate in cell adhesion to fibrinogen, a common ligand of CR3 and CR4, employing human monocytes, monocyte-derived macrophages (MDMs) and monocyte-derived dendritic cells (MDDCs) highly expressing CD11b as well as CD11c. We determined the exact numbers of CD11b and CD11c on these cell types by a bead-based technique, and found that the ratio of CD11b/CD11c is 1.2 for MDDCs, 1.7 for MDMs and 7.1 for monocytes, suggesting that the function of CD11c is preponderant in MDDCs and less pronounced in monocytes. Applying state-of-the-art biophysical techniques, we proved that cellular adherence to fibrinogen is dominated by CD11c. Furthermore, we found that blocking CD11b significantly enhances the attachment of MDDCs and MDMs to fibrinogen, demonstrating a competition between CD11b and CD11c for this ligand. On the basis of the cell surface receptor numbers and the measured adhesion strength we set up a model, which explains the different behavior of the three cell types.
Collapse
Affiliation(s)
- Noémi Sándor
- MTA-ELTE Immunology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Szilvia Lukácsi
- Department of Immunology, Institute of Biology, Faculty of Science, Eötvös Loránd University, Budapest, Hungary
| | - Rita Ungai-Salánki
- Department of Biological Physics, Institute of Physics, Faculty of Science, Eötvös Loránd University, Budapest, Hungary
| | - Norbert Orgován
- Nanobiosensorics “Lendület” Group, Institute of Technical Physics and Material Sciences, Centre for Energy Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Bálint Szabó
- Department of Biological Physics, Institute of Physics, Faculty of Science, Eötvös Loránd University, Budapest, Hungary
| | - Róbert Horváth
- Nanobiosensorics “Lendület” Group, Institute of Technical Physics and Material Sciences, Centre for Energy Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Anna Erdei
- MTA-ELTE Immunology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
- Department of Immunology, Institute of Biology, Faculty of Science, Eötvös Loránd University, Budapest, Hungary
| | - Zsuzsa Bajtay
- Department of Immunology, Institute of Biology, Faculty of Science, Eötvös Loránd University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
27
|
Prostate-specific membrane antigen (PSMA)-mediated laminin proteolysis generates a pro-angiogenic peptide. Angiogenesis 2016; 19:487-500. [PMID: 27387982 DOI: 10.1007/s10456-016-9521-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/21/2016] [Indexed: 02/06/2023]
Abstract
Prostate-specific membrane antigen (PSMA) is a membrane-bound glutamate carboxypeptidase expressed in a number of tissues. PSMA participates in various biological functions depending on the substrate available in the particular tissue; in the brain, PSMA cleaves the abundant neuropeptide N-acetyl-aspartyl-glutamate to regulate release of key neurotransmitters, while intestinal PSMA cleaves polyglutamated peptides to supply dietary folate. PSMA expression is also progressively upregulated in prostate cancer where it correlates with tumor progression as well as in tumor vasculature, where it regulates angiogenesis. The previous research determined that PSMA cleavage of small peptides generated via matrix metalloprotease-mediated proteolysis of the extracellular matrix protein laminin potently activated endothelial cells, integrin signaling and angiogenesis, although the specific peptide substrates were not identified. Herein, using enzymatic analyses and LC/MS, we unequivocally demonstrate that several laminin-derived peptides containing carboxy-terminal glutamate moieties (LQE, IEE, LNE) are bona fide substrates for PSMA. Subsequently, the peptide products were tested for their effects on angiogenesis in various models. We report that LQ, the dipeptide product of PSMA cleavage of LQE, efficiently activates endothelial cells in vitro and enhances angiogenesis in vivo. Importantly, LQE is not cleaved by an inactive PSMA enzyme containing an active site mutation (E424S). Endothelial cell activation by LQ was dependent on integrin beta-1-induced activation of focal adhesion kinase. These results characterize a novel PSMA substrate, provide a functional rationale for the upregulation of PSMA in cancer cells and tumor vasculature and suggest that inhibition of PSMA could lead to the development of new angiogenic therapies.
Collapse
|
28
|
Jensen MR, Bajic G, Zhang X, Laustsen AK, Koldsø H, Skeby KK, Schiøtt B, Andersen GR, Vorup-Jensen T. Structural Basis for Simvastatin Competitive Antagonism of Complement Receptor 3. J Biol Chem 2016; 291:16963-76. [PMID: 27339893 DOI: 10.1074/jbc.m116.732222] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Indexed: 01/08/2023] Open
Abstract
The complement system is an important part of the innate immune response to infection but may also cause severe complications during inflammation. Small molecule antagonists to complement receptor 3 (CR3) have been widely sought, but a structural basis for their mode of action is not available. We report here on the structure of the human CR3 ligand-binding I domain in complex with simvastatin. Simvastatin targets the metal ion-dependent adhesion site of the open, ligand-binding conformation of the CR3 I domain by direct contact with the chelated Mg(2+) ion. Simvastatin antagonizes I domain binding to the complement fragments iC3b and C3d but not to intercellular adhesion molecule-1. By virtue of the I domain's wide distribution in binding kinetics to ligands, it was possible to identify ligand binding kinetics as discriminator for simvastatin antagonism. In static cellular experiments, 15-25 μm simvastatin reduced adhesion by K562 cells expressing recombinant CR3 and by primary human monocytes, with an endogenous expression of this receptor. Application of force to adhering monocytes potentiated the effects of simvastatin where only a 50-100 nm concentration of the drug reduced the adhesion by 20-40% compared with untreated cells. The ability of simvastatin to target CR3 in its ligand binding-activated conformation is a novel mechanism to explain the known anti-inflammatory effects of this compound, in particular because this CR3 conformation is found in pro-inflammatory environments. Our report points to new designs of CR3 antagonists and opens new perspectives and identifies druggable receptors from characterization of the ligand binding kinetics in the presence of antagonists.
Collapse
Affiliation(s)
| | - Goran Bajic
- Molecular Biology and Genetics, and the Lundbeck Foundation Nanomedicine Center for Individualized Management of Tissue Damage and Regeneration (LUNA), and
| | | | | | - Heidi Koldsø
- Chemistry, the Interdisciplinary Nanoscience Center (iNANO), the Center for Insoluble Protein Structures (inSPIN)
| | - Katrine Kirkeby Skeby
- Chemistry, the Interdisciplinary Nanoscience Center (iNANO), the Center for Insoluble Protein Structures (inSPIN)
| | - Birgit Schiøtt
- Chemistry, the Interdisciplinary Nanoscience Center (iNANO), the Center for Insoluble Protein Structures (inSPIN)
| | - Gregers R Andersen
- Molecular Biology and Genetics, and the Lundbeck Foundation Nanomedicine Center for Individualized Management of Tissue Damage and Regeneration (LUNA), and
| | - Thomas Vorup-Jensen
- From the Departments of Biomedicine, the Lundbeck Foundation Nanomedicine Center for Individualized Management of Tissue Damage and Regeneration (LUNA), and the Interdisciplinary Nanoscience Center (iNANO), the MEMBRANES Research Center, Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
29
|
Expression of CD11c Is Associated with Unconventional Activated T Cell Subsets with High Migratory Potential. PLoS One 2016; 11:e0154253. [PMID: 27119555 PMCID: PMC4847787 DOI: 10.1371/journal.pone.0154253] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 04/11/2016] [Indexed: 11/19/2022] Open
Abstract
CD11c is an α integrin classically employed to define myeloid dendritic cells. Although there is little information about CD11c expression on human T cells, mouse models have shown an association of CD11c expression with functionally relevant T cell subsets. In the context of genital tract infection, we have previously observed increased expression of CD11c in circulating T cells from mice and women. Microarray analyses of activated effector T cells expressing CD11c derived from naïve mice demonstrated enrichment for natural killer (NK) associated genes. Here we find that murine CD11c+ T cells analyzed by flow cytometry display markers associated with non-conventional T cell subsets, including γδ T cells and invariant natural killer T (iNKT) cells. However, in women, only γδ T cells and CD8+ T cells were enriched within the CD11c fraction of blood and cervical tissue. These CD11c+ cells were highly activated and had greater interferon (IFN)-γ secretory capacity than CD11c- T cells. Furthermore, circulating CD11c+ T cells were associated with the expression of multiple adhesion molecules in women, suggesting that these cells have high tissue homing potential. These data suggest that CD11c expression distinguishes a population of circulating T cells during bacterial infection with innate capacity and mucosal homing potential.
Collapse
|
30
|
Li H, Borrego F, Nagata S, Tolnay M. Fc Receptor-like 5 Expression Distinguishes Two Distinct Subsets of Human Circulating Tissue-like Memory B Cells. THE JOURNAL OF IMMUNOLOGY 2016; 196:4064-74. [PMID: 27076679 DOI: 10.4049/jimmunol.1501027] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 02/29/2016] [Indexed: 12/22/2022]
Abstract
Fc receptor-like (FCRL) 5 is a novel IgG binding protein expressed on B cells, with the capacity to regulate Ag receptor signaling. We assessed FCRL5 expression on circulating B cells from healthy donors and found that FCRL5(+) cells are most enriched among atypical CD21(-/lo)/CD27(-) tissue-like memory (TLM) B cells, which are abnormally expanded in several autoimmune and infectious diseases. Using multicolor flow cytometry, FCRL5(+) TLM cells were found to express more CD11c and several inhibitory receptors than did the FCRL5(-) TLM subset. The homing receptor profiles of the two TLM subsets shared features consistent with migration away from lymphoid tissues, but they also displayed distinct differences. Analysis of IgH V regions in single cells indicated that although both subsets are diverse, the FCRL5(+) subset accumulated significantly more somatic mutations. Furthermore, the FCRL5(+) subset had more switched isotype expression and more extensive proliferative history. Microarray analysis and quantitative RT-PCR demonstrated that the two TLM subsets possess distinct gene expression profiles, characterized by markedly different CD11c, SOX5, T-bet, and RTN4R expression, as well as differences in expression of inhibitory receptors. Functional analysis revealed that the FCRL5(+) TLM subset responds poorly to multiple stimuli compared with the FCRL5(-) subset, as reflected by reduced calcium mobilization and blunted cell proliferation. We propose that the FCRL5(+) TLM subset, but not the FCRL5(-) TLM subset, underwent Ag-driven development and is severely dysfunctional. The present study elucidates the heterogeneity of TLM B cells and provides the basis to dissect their roles in the pathogenesis of inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Huifang Li
- Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - Francisco Borrego
- Immunopathology Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain; and
| | - Satoshi Nagata
- Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Mate Tolnay
- Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993;
| |
Collapse
|
31
|
Zhang X, Bajic G, Andersen GR, Christiansen SH, Vorup-Jensen T. The cationic peptide LL-37 binds Mac-1 (CD11b/CD18) with a low dissociation rate and promotes phagocytosis. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:471-8. [PMID: 26876535 DOI: 10.1016/j.bbapap.2016.02.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/03/2016] [Accepted: 02/10/2016] [Indexed: 12/16/2022]
Abstract
As a broad-spectrum anti-microbial peptide, LL-37 plays an important role in the innate immune system. A series of previous reports implicates LL-37 as an activator of various cell surface receptor-mediated functions, including chemotaxis in integrin CD11b/CD18 (Mac-1)-expressing cells. However, evidence is scarce concerning the direct binding of LL-37 to these receptors and investigations on the associated binding kinetics is lacking. Mac-1, a member of the β2 integrin family, is mainly expressed in myeloid leukocytes. Its critical functions include phagocytosis of complement-opsonized pathogens. Here, we report on interactions of LL-37 and its fragment FK-13 with the ligand-binding domain of Mac-1, the α-chain I domain. LL-37 bound the I-domain with an affinity comparable to the complement fragment C3d, one of the strongest known ligands for Mac-1. In cell adhesion assays both LL-37 and FK-13 supported binding by Mac-1 expressing cells, however, with LL-37-coupled surfaces supporting stronger cell adhesion than FK-13. Likewise, in phagocytosis assays with primary human monocytes both LL-37 and FK-13 enhanced uptake of particles coupled with these ligands but with a tendency towards a stronger uptake by LL-37.
Collapse
Affiliation(s)
- Xianwei Zhang
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Goran Bajic
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Gregers R Andersen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark
| | | | - Thomas Vorup-Jensen
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark; MEMBRANES Research Center, Aarhus University, DK-8000 Aarhus C, Denmark.
| |
Collapse
|
32
|
Innate Immunity and Biomaterials at the Nexus: Friends or Foes. BIOMED RESEARCH INTERNATIONAL 2015; 2015:342304. [PMID: 26247017 PMCID: PMC4515263 DOI: 10.1155/2015/342304] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 06/15/2015] [Accepted: 06/22/2015] [Indexed: 01/04/2023]
Abstract
Biomaterial implants are an established part of medical practice, encompassing a broad range of devices that widely differ in function and structural composition. However, one common property amongst biomaterials is the induction of the foreign body response: an acute sterile inflammatory reaction which overlaps with tissue vascularisation and remodelling and ultimately fibrotic encapsulation of the biomaterial to prevent further interaction with host tissue. Severity and clinical manifestation of the biomaterial-induced foreign body response are different for each biomaterial, with cases of incompatibility often associated with loss of function. However, unravelling the mechanisms that progress to the formation of the fibrotic capsule highlights the tightly intertwined nature of immunological responses to a seemingly noncanonical “antigen.” In this review, we detail the pathways associated with the foreign body response and describe possible mechanisms of immune involvement that can be targeted. We also discuss methods of modulating the immune response by altering the physiochemical surface properties of the biomaterial prior to implantation. Developments in these areas are reliant on reproducible and effective animal models and may allow a “combined” immunomodulatory approach of adapting surface properties of biomaterials, as well as treating key immune pathways to ultimately reduce the negative consequences of biomaterial implantation.
Collapse
|
33
|
Kläning E, Christensen B, Bajic G, Hoffmann SV, Jones NC, Callesen MM, Andersen GR, Sørensen ES, Vorup-Jensen T. Multiple low-affinity interactions support binding of human osteopontin to integrin αXβ2. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:930-8. [PMID: 25839998 DOI: 10.1016/j.bbapap.2015.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 03/18/2015] [Accepted: 03/22/2015] [Indexed: 01/11/2023]
Abstract
Integrin α(X)β(2) (also known as complement receptor 4, p150,95, or CD11c/CD18) is expressed in the cell membrane of myeloid leukocytes. α(X)β(2) has been reported to bind a large number of structurally unrelated ligands, often with a shared molecular character in the presence of polyanionic stretches in poorly folded proteins or glucosaminoglycans. Nevertheless, it is unclear what chemical sources of polyanionicity enable the binding by α(X)β(2). Osteopontin (OPN) is an intrinsically disordered protein, which facilitates phagocytosis via the integrin α(X)β(2). Unlike for other integrins, neither the RGD nor the SVVYGLR motifs account for this binding, and the molecular basis of OPN binding by α(X)β(2) remains uncharacterized. Here, we show that the monovalent interactions between the ligand-binding domain of α(X)β(2) and OPN, its fragments, or caseins are weak, with dissociation constants higher than 10(-5)M but with high apparent stoichiometries. From comparison with cell adhesion studies, the discrimination between α(X)β(2) ligands and non-ligands appears to rely on these apparent stoichiometries in a way, which involves glutamate rather than aspartate side chains. Surprisingly, the extensive, negatively charged phosphorylation of OPN is not contributing to α(X)β(2) binding. Furthermore, synchrotron radiation circular spectroscopy excludes that the phosphorylation affects the general folding of OPN. Taken together, our quantitative analyses reveal a mode of ligand recognition by integrin α(X)β(2), which seem to differ in principles considerably from other OPN receptors.
Collapse
Affiliation(s)
- Eva Kläning
- Dept. of Molecular Biology and Genetics Aarhus University, Aarhus, Denmark; Dept. of Biomedicine, Denmark
| | - Brian Christensen
- Dept. of Molecular Biology and Genetics Aarhus University, Aarhus, Denmark
| | - Goran Bajic
- Dept. of Molecular Biology and Genetics Aarhus University, Aarhus, Denmark
| | - Søren V Hoffmann
- Institute for Storage Ring Facilities Aarhus (ISA), Dept. of Physics and Astronomy & Center for Storage Ring Facilities Aarhus, Denmark
| | - Nykola C Jones
- Institute for Storage Ring Facilities Aarhus (ISA), Dept. of Physics and Astronomy & Center for Storage Ring Facilities Aarhus, Denmark
| | - Morten M Callesen
- Dept. of Molecular Biology and Genetics Aarhus University, Aarhus, Denmark
| | - Gregers R Andersen
- Dept. of Molecular Biology and Genetics Aarhus University, Aarhus, Denmark
| | - Esben S Sørensen
- Dept. of Molecular Biology and Genetics Aarhus University, Aarhus, Denmark; Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus Denmark
| | - Thomas Vorup-Jensen
- Dept. of Biomedicine, Denmark; Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus Denmark; MEMBRANES Research Center, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
34
|
Steblyanko M, Anikeeva N, Campbell KS, Keen JH, Sykulev Y. Integrins Influence the Size and Dynamics of Signaling Microclusters in a Pyk2-dependent Manner. J Biol Chem 2015; 290:11833-42. [PMID: 25778396 DOI: 10.1074/jbc.m114.614719] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Indexed: 12/28/2022] Open
Abstract
Integrin engagement on lymphocytes initiates "outside-in" signaling that is required for cytoskeleton remodeling and the formation of the synaptic interface. However, the mechanism by which the "outside-in" signal contributes to receptor-mediated intracellular signaling that regulates the kinetics of granule delivery and efficiency of cytolytic activity is not well understood. We have found that variations in ICAM-1 expression on tumor cells influence killing kinetics of these cells by CD16.NK-92 cytolytic effectors suggesting that changes in integrin ligation on the effector cells regulate the kinetics of cytolytic activity by the effector cells. To understand how variations of the integrin receptor ligation may alter cytolytic activity of CD16.NK-92 cells, we analyzed molecular events at the contact area of these cells exposed to planar lipid bilayers that display integrin ligands at different densities and activating CD16-specific antibodies. Changes in the extent of integrin ligation on CD16.NK-92 cells at the cell/bilayer interface revealed that the integrin signal influences the size and the dynamics of activating receptor microclusters in a Pyk2-dependent manner. Integrin-mediated changes of the intracellular signaling significantly affected the kinetics of degranulation of CD16.NK-92 cells providing evidence that integrins regulate the rate of target cell destruction in antibody-dependent cell cytotoxicity (ADCC).
Collapse
Affiliation(s)
| | | | - Kerry S Campbell
- the Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | - James H Keen
- Biochemistry and Molecular Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| | - Yuri Sykulev
- From the Departments of Microbiology and Immunology and
| |
Collapse
|
35
|
Cromvik J, Johnsson M, Vaht K, Johansson JE, Wennerås C. Eosinophils in the blood of hematopoietic stem cell transplanted patients are activated and have different molecular marker profiles in acute and chronic graft-versus-host disease. Immun Inflamm Dis 2014; 2:99-113. [PMID: 25400930 PMCID: PMC4217552 DOI: 10.1002/iid3.25] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/14/2014] [Accepted: 05/16/2014] [Indexed: 12/20/2022] Open
Abstract
While increased numbers of eosinophils may be detected in patients with graft-versus-host disease (GVHD) following hematopoietic stem cell transplantation, it is not known if eosinophils play a role in GVHD. The aims of this study were to determine: whether eosinophils are activated during GVHD; whether the patterns of activation are similar in acute and chronic GVHD; and the ways in which systemic corticosteroids affect eosinophils. Transplanted patients (n = 35) were investigated for eosinophil numbers and the expression levels of 16 eosinophilic cell surface markers using flow cytometry; all the eosinophil data were analyzed by the multivariate method OPLS-DA. Different patterns of molecule expression were observed on the eosinophils from patients with acute, chronic, and no GVHD, respectively. The molecules that provided the best discrimination between acute and chronic GVHD were: the activation marker CD9; adhesion molecules CD11c and CD18; chemokine receptor CCR3; and prostaglandin receptor CRTH2. Patients with acute or chronic GVHD who received systemic corticosteroid treatment showed down-regulation of the cell surface markers on their eosinophils, whereas corticosteroid treatment had no effect on the eosinophil phenotype in the patients without GVHD. In summary, eosinophils are activated in GVHD, display different activation profiles in acute and chronic GVHD, and are highly responsive to systemic corticosteroids.
Collapse
Affiliation(s)
- Julia Cromvik
- Department of Hematology and Coagulation, University of Gothenburg Göteborg, Sweden
| | - Marianne Johnsson
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg Göteborg, Sweden
| | - Krista Vaht
- Department of Hematology and Coagulation, University of Gothenburg Göteborg, Sweden
| | - Jan-Erik Johansson
- Department of Hematology and Coagulation, University of Gothenburg Göteborg, Sweden
| | - Christine Wennerås
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg Göteborg, Sweden
| |
Collapse
|
36
|
Raftery MJ, Lalwani P, Krautkrӓmer E, Peters T, Scharffetter-Kochanek K, Krüger R, Hofmann J, Seeger K, Krüger DH, Schönrich G. β2 integrin mediates hantavirus-induced release of neutrophil extracellular traps. ACTA ACUST UNITED AC 2014; 211:1485-97. [PMID: 24889201 PMCID: PMC4076588 DOI: 10.1084/jem.20131092] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
β2 Integrin–mediated systemic release of neutrophil extracellular traps is a novel mechanism of immunopathology associated with hantavirus infection. Rodent-borne hantaviruses are emerging human pathogens that cause severe human disease. The underlying mechanisms are not well understood, as hantaviruses replicate in endothelial and epithelial cells without causing any cytopathic effect. We demonstrate that hantaviruses strongly stimulated neutrophils to release neutrophil extracellular traps (NETs). Hantavirus infection induced high systemic levels of circulating NETs in patients and this systemic NET overflow was accompanied by production of autoantibodies to nuclear antigens. Analysis of the responsible mechanism using neutrophils from β2 null mice identified β2 integrin receptors as a master switch for NET induction. Further experiments suggested that β2 integrin receptors such as complement receptor 3 (CR3) and 4 (CR4) may act as novel hantavirus entry receptors. Using adenoviruses, we confirmed that viral interaction with β2 integrin induced strong NET formation. Collectively, β2 integrin–mediated systemic NET overflow is a novel viral mechanism of immunopathology that may be responsible for characteristic aspects of hantavirus-associated disease such as kidney and lung damage.
Collapse
Affiliation(s)
- Martin J Raftery
- Institute of Medical Virology, Helmut-Ruska-Haus, Department of Pediatric Pneumology and Immunology, and Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Pritesh Lalwani
- Institute of Medical Virology, Helmut-Ruska-Haus, Department of Pediatric Pneumology and Immunology, and Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Ellen Krautkrӓmer
- Department of Nephrology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Thorsten Peters
- Department of Dermatology and Allergic Diseases, University Hospital Ulm, 89081 Ulm, Germany
| | | | - Renate Krüger
- Institute of Medical Virology, Helmut-Ruska-Haus, Department of Pediatric Pneumology and Immunology, and Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Jörg Hofmann
- Institute of Medical Virology, Helmut-Ruska-Haus, Department of Pediatric Pneumology and Immunology, and Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany Division of Virology, Labor Berlin Charité-Vivantes GmbH, 13353 Berlin, Germany
| | - Karl Seeger
- Institute of Medical Virology, Helmut-Ruska-Haus, Department of Pediatric Pneumology and Immunology, and Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Detlev H Krüger
- Institute of Medical Virology, Helmut-Ruska-Haus, Department of Pediatric Pneumology and Immunology, and Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Günther Schönrich
- Institute of Medical Virology, Helmut-Ruska-Haus, Department of Pediatric Pneumology and Immunology, and Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
37
|
Gorudko IV, Grigorieva DV, Shamova EV, Kostevich VA, Sokolov AV, Mikhalchik EV, Cherenkevich SN, Arnhold J, Panasenko OM. Hypohalous acid-modified human serum albumin induces neutrophil NADPH oxidase activation, degranulation, and shape change. Free Radic Biol Med 2014; 68:326-34. [PMID: 24384524 DOI: 10.1016/j.freeradbiomed.2013.12.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 12/18/2013] [Accepted: 12/20/2013] [Indexed: 12/14/2022]
Abstract
Halogenated lipids, proteins, and lipoproteins formed in reactions with myeloperoxidase (MPO)-derived hypochlorous acid (HOCl) and hypobromous acid (HOBr) can contribute to the regulation of functional activity of cells and serve as mediators of inflammation. Human serum albumin (HSA) is the major plasma protein target of hypohalous acids. This study was performed to assess the potency of HSA modified by HOCl (HSA-Cl) and HOBr (HSA-Br) to elicit selected neutrophil responses. HSA-Cl/Br were found to induce neutrophil degranulation, generation of reactive oxygen intermediates, shape change, and actin cytoskeleton reorganization. Thus HSA-Cl/Br can initially act as a switch and then as a feeder of the "inflammatory loop" under oxidative stress. In HSA-Cl/Br-treated neutrophils, monoclonal antibodies against CD18, the β subunit of β2 integrins, reduced the production of superoxide anion radicals and hydrogen peroxide as well as MPO exocytosis, suggesting that CD18 contributed to neutrophil activation. HSA-Cl/Br-induced neutrophil responses were also inhibited by genistein, a broad-specificity tyrosine kinase inhibitor, and wortmannin, a phosphoinositide 3-kinase (PI3K) inhibitor, supporting the notion that activation of both tyrosine kinase and PI3K may play a role in neutrophil activation by HSA modified in MPO-dependent reactions. These results confirm the hypothesis that halogenated molecules formed in vivo via MPO-dependent reactions can be considered as a new class of biologically active substances potentially able to contribute to activation of myeloid cells in sites of inflammation and serve as inflammatory response modulators.
Collapse
Affiliation(s)
- Irina V Gorudko
- Department of Biophysics, Belarusian State University, Minsk 220050, Belarus.
| | - Daria V Grigorieva
- Department of Biophysics, Belarusian State University, Minsk 220050, Belarus
| | - Ekaterina V Shamova
- Department of Biophysics, Belarusian State University, Minsk 220050, Belarus
| | - Valeria A Kostevich
- Institute of Experimental Medicine, Saint-Petersburg 197376, Russia; Research Institute of Physico-Chemical Medicine, Moscow 119435, Russia
| | - Alexey V Sokolov
- Institute of Experimental Medicine, Saint-Petersburg 197376, Russia; Research Institute of Physico-Chemical Medicine, Moscow 119435, Russia; State University of Saint Petersburg, Saint Petersburg 199000, Russia
| | | | | | - Jürgen Arnhold
- Institute for Medical Physics and Biophysics, Medical Faculty, University of Leipzig, 04107 Leipzig, Germany
| | - Oleg M Panasenko
- Research Institute of Physico-Chemical Medicine, Moscow 119435, Russia
| |
Collapse
|
38
|
Abstract
In the cell membrane complement receptor 3 (CR3) consists of one alpha chain (CD11b) and one beta chain (CD18). CR3 participates in many immunological processes, especially those involving cell migration, adhesion, and phagocytosis of complement-opsonized microbes. Recent findings of soluble CR3 in body fluids and in culture supernatant from experiments in vitro point to the involvement of ecto domain shedding as a part of the CR3 biology. To monitor such shedding on a quantitative basis, we have developed time-resolved immunofluorometric assays (TRIFMA) to detect soluble CD11b and CD18 in plasma or serum of either human or murine origin. Compared with most enzyme-linked immunosorbent assays methodologies, TRIFMA possesses prominent advantages, including better dynamic range and reproducibility. These assays may contribute to the understanding of the role of shedding of CR3 and other cell adhesion molecules in human disease and animal models involving inflammation.
Collapse
|
39
|
Ballut L, Sapay N, Chautard E, Imberty A, Ricard-Blum S. Mapping of heparin/heparan sulfate binding sites on αvβ3 integrin by molecular docking. J Mol Recognit 2013; 26:76-85. [PMID: 23334915 DOI: 10.1002/jmr.2250] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 10/15/2012] [Accepted: 10/15/2012] [Indexed: 01/24/2023]
Abstract
Heparin/heparan sulfate interact with growth factors, chemokines, extracellular proteins, and receptors. Integrins are αβ heterodimers that serve as receptors for extracellular proteins, regulate cell behavior, and participate in extracellular matrix assembly. Heparin binds to RGD-dependent integrins (αIIbβ3, α5β1, αvβ3, and αvβ5) and to RGD-independent integrins (α4β1, αXβ2, and αMβ2), but their binding sites have not been located on integrins. We report the mapping of heparin binding sites on the ectodomain of αvβ3 integrin by molecular modeling. The surface of the ectodomain was scanned with small rigid probes mimicking the sulfated domains of heparan sulfate. Docking results were clustered into binding spots. The best results were selected for further docking simulations with heparin hexasaccharide. Six potential binding spots containing lysine and/or arginine residues were identified on the ectodomain of αvβ3 integrin. Heparin would mostly bind to the top of the genu domain, the Calf-I domain of the α subunit, and the top of the β subunit of RGD-dependent integrins. Three spots were close enough from each other on the integrin surface to form an extended binding site that could interact with heparin/heparan sulfate chains. Because heparin does not bind to the same integrin site as protein ligands, no steric hindrance prevents the formation of ternary complexes comprising the integrin, its protein ligand, and heparin/heparan sulfate. The basic amino acid residues predicted to interact with heparin are conserved in the sequences of RGD-dependent but not of RGD-independent integrins suggesting that heparin/heparan sulfate could bind to different sites on these two integrin subfamilies.
Collapse
Affiliation(s)
- Lionel Ballut
- UMR 5086 CNRS-Université Lyon 1, Institut de Biologie et Chimie des Protéines, 7 passage du Vercors, 69367 Lyon Cedex 07, France
| | | | | | | | | |
Collapse
|
40
|
Jalilian B, Christiansen SH, Einarsson HB, Pirozyan MR, Petersen E, Vorup-Jensen T. Properties and prospects of adjuvants in influenza vaccination - messy precipitates or blessed opportunities? MOLECULAR AND CELLULAR THERAPIES 2013; 1:2. [PMID: 26056568 PMCID: PMC4448954 DOI: 10.1186/2052-8426-1-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 09/10/2013] [Indexed: 01/09/2023]
Abstract
Influenza is a major challenge to healthcare systems world-wide. While prophylactic vaccination is largely efficient, long-lasting immunity has not been achieved in immunized populations, at least in part due to the challenges arising from the antigen variation between strains of influenza A virus as a consequence of genetic drift and shift. From progress in our understanding of the immune system, the mode-of-action of vaccines can be divided into the stimulation of the adaptive system through inclusion of appropriate vaccine antigens and of the innate immune system by the addition of adjuvant to the vaccine formulation. A shared property of many vaccine adjuvants is found in their nature of water-insoluble precipitates, for instance the particulate material made from aluminum salts. Previously, it was thought that embedding of vaccine antigens in these materials provided a "depot" of antigens enabling a long exposure of the immune system to the antigen. However, more recent work points to a role of particulate adjuvants in stimulating cellular parts of the innate immune system. Here, we briefly outline the infectious medicine and immune biology of influenza virus infection and procedures to provide sufficient and stably available amounts of vaccine antigen. This is followed by presentation of the many roles of adjuvants, which involve humoral factors of innate immunity, notably complement. In a perspective of the ultrastructural properties of these humoral factors, it becomes possible to rationalize why these insoluble precipitates or emulsions are such a provocation of the immune system. We propose that the biophysics of particulate material may hold opportunities that could aid the development of more efficient influenza vaccines.
Collapse
Affiliation(s)
- Babak Jalilian
- Biophysical Immunology Laboratory, Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| | - Stig Hill Christiansen
- Biophysical Immunology Laboratory, Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| | - Halldór Bjarki Einarsson
- Biophysical Immunology Laboratory, Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark ; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mehdi Rasoli Pirozyan
- Inflammation and Infection Research Centre, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Eskild Petersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark ; Department of Infectious Medicine (Q), Aarhus University Hospital, Aarhus, Denmark
| | - Thomas Vorup-Jensen
- Biophysical Immunology Laboratory, Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| |
Collapse
|
41
|
Jalilian B, Christiansen SH, Einarsson HB, Pirozyan MR, Petersen E, Vorup-Jensen T. Properties and prospects of adjuvants in influenza vaccination - messy precipitates or blessed opportunities? MOLECULAR AND CELLULAR THERAPIES 2013; 1:2. [PMID: 26056568 PMCID: PMC4448954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 09/10/2013] [Indexed: 11/21/2023]
Abstract
Influenza is a major challenge to healthcare systems world-wide. While prophylactic vaccination is largely efficient, long-lasting immunity has not been achieved in immunized populations, at least in part due to the challenges arising from the antigen variation between strains of influenza A virus as a consequence of genetic drift and shift. From progress in our understanding of the immune system, the mode-of-action of vaccines can be divided into the stimulation of the adaptive system through inclusion of appropriate vaccine antigens and of the innate immune system by the addition of adjuvant to the vaccine formulation. A shared property of many vaccine adjuvants is found in their nature of water-insoluble precipitates, for instance the particulate material made from aluminum salts. Previously, it was thought that embedding of vaccine antigens in these materials provided a "depot" of antigens enabling a long exposure of the immune system to the antigen. However, more recent work points to a role of particulate adjuvants in stimulating cellular parts of the innate immune system. Here, we briefly outline the infectious medicine and immune biology of influenza virus infection and procedures to provide sufficient and stably available amounts of vaccine antigen. This is followed by presentation of the many roles of adjuvants, which involve humoral factors of innate immunity, notably complement. In a perspective of the ultrastructural properties of these humoral factors, it becomes possible to rationalize why these insoluble precipitates or emulsions are such a provocation of the immune system. We propose that the biophysics of particulate material may hold opportunities that could aid the development of more efficient influenza vaccines.
Collapse
Affiliation(s)
- Babak Jalilian
- />Biophysical Immunology Laboratory, Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| | - Stig Hill Christiansen
- />Biophysical Immunology Laboratory, Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| | - Halldór Bjarki Einarsson
- />Biophysical Immunology Laboratory, Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
- />Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mehdi Rasoli Pirozyan
- />Inflammation and Infection Research Centre, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Eskild Petersen
- />Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- />Department of Infectious Medicine (Q), Aarhus University Hospital, Aarhus, Denmark
| | - Thomas Vorup-Jensen
- />Biophysical Immunology Laboratory, Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| |
Collapse
|
42
|
Uotila LM, Aatonen M, Gahmberg CG. Integrin CD11c/CD18 α-chain phosphorylation is functionally important. J Biol Chem 2013; 288:33494-9. [PMID: 24129562 DOI: 10.1074/jbc.c113.497446] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CD11c/CD18 (αXβ2, p150/95, or complement receptor 4, CR4) is a monocyte/macrophage-enriched integrin that has been reported to bind to a variety of ligands. These include cell surface proteins, extracellular matrix proteins, and soluble ligands. The regulation of ligand binding to CD11c/CD18 has remained poorly understood. Previous work has shown that both α-chain and β-chain phosphorylations of CD11a/CD18 and CD11b/CD18 are needed for activity, but no corresponding studies on CD11c/CD18 have been performed. In this study, we have identified the phosphorylation site of CD11c as Ser-1158 and show that it is pivotal for adherence and phagocytosis.
Collapse
Affiliation(s)
- Liisa M Uotila
- From the Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland
| | | | | |
Collapse
|
43
|
Structural insight on the recognition of surface-bound opsonins by the integrin I domain of complement receptor 3. Proc Natl Acad Sci U S A 2013; 110:16426-31. [PMID: 24065820 DOI: 10.1073/pnas.1311261110] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Complement receptors (CRs), expressed notably on myeloid and lymphoid cells, play an essential function in the elimination of complement-opsonized pathogens and apoptotic/necrotic cells. In addition, these receptors are crucial for the cross-talk between the innate and adaptive branches of the immune system. CR3 (also known as Mac-1, integrin αMβ2, or CD11b/CD18) is expressed on all macrophages and recognizes iC3b on complement-opsonized objects, enabling their phagocytosis. We demonstrate that the C3d moiety of iC3b harbors the binding site for the CR3 αI domain, and our structure of the C3d:αI domain complex rationalizes the CR3 selectivity for iC3b. Based on extensive structural analysis, we suggest that the choice between a ligand glutamate or aspartate for coordination of a receptor metal ion-dependent adhesion site-bound metal ion is governed by the secondary structure of the ligand. Comparison of our structure to the CR2:C3d complex and the in vitro formation of a stable CR3:C3d:CR2 complex suggests a molecular mechanism for the hand-over of CR3-bound immune complexes from macrophages to CR2-presenting cells in lymph nodes.
Collapse
|
44
|
Shape change in the receptor for gliding motility in Plasmodium sporozoites. Proc Natl Acad Sci U S A 2012; 109:21420-5. [PMID: 23236185 DOI: 10.1073/pnas.1218581109] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Sporozoite gliding motility and invasion of mosquito and vertebrate host cells in malaria is mediated by thrombospondin repeat anonymous protein (TRAP). Tandem von Willebrand factor A (VWA) and thrombospondin type I repeat (TSR) domains in TRAP connect through proline-rich stalk, transmembrane, and cytoplasmic domains to the parasite actin-dependent motility apparatus. We crystallized fragments containing the VWA and TSR domains from Plasmodium vivax and Plasmodium falciparum in different crystal lattices. TRAP VWA domains adopt closed and open conformations, and bind a Mg(2+) ion at a metal ion-dependent adhesion site implicated in ligand binding. Metal ion coordination in the open state is identical to that seen in the open high-affinity state of integrin I domains. The closed VWA conformation associates with a disordered TSR domain. In contrast, the open VWA conformation crystallizes with an extensible β ribbon and ordered TSR domain. The extensible β ribbon is composed of disulfide-bonded segments N- and C-terminal to the VWA domain that are largely drawn out of the closed VWA domain in a 15 Å movement to the open conformation. The extensible β ribbon and TSR domain overlap at a conserved interface. The VWA, extensible β ribbon, and TSR domains adopt a highly elongated overall orientation that would be stabilized by tensile force exerted across a ligand-receptor complex by the actin motility apparatus of the sporozoite. Our results provide insights into regulation of "stick-and-slip" parasite motility and for development of sporozoite subunit vaccines.
Collapse
|
45
|
Reinholdt J, Poulsen K, Brinkmann CR, Hoffmann SV, Stapulionis R, Enghild JJ, Jensen UB, Boesen T, Vorup-Jensen T. Monodisperse and LPS-free Aggregatibacter actinomycetemcomitans leukotoxin: interactions with human β2 integrins and erythrocytes. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2012; 1834:546-58. [PMID: 23234758 DOI: 10.1016/j.bbapap.2012.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 11/15/2012] [Accepted: 12/03/2012] [Indexed: 01/08/2023]
Abstract
Aggregatibacter actinomycetemcomitans is a gram-negative, facultatively anaerobic cocco-bacillus and a frequent member of the human oral flora. It produces a leukotoxin, LtxA, belonging to the repeats-in-toxin (RTX) family of bacterial cytotoxins. LtxA efficiently kills neutrophils and mononuclear phagocytes. The known receptor for LtxA on leukocytes is integrin α(L)β(2) (LFA-1 or CD11a/CD18). However, the molecular mechanisms involved in LtxA-mediated cytotoxicity are poorly understood, partly because LtxA has proven difficult to prepare for experiments as free of contaminants and with its native structure. Here, we describe a protocol for the purification of LtxA from bacterial culture supernatant, which does not involve denaturing procedures. The purified LtxA was monodisperse, well folded as judged by the combined use of synchrotron radiation circular dichroism spectroscopy (SRCD) and in silico prediction of the secondary structure content, and free of bacterial lipopolysaccharide. The analysis by SRCD and similarity to a lipase from Pseudomonas with a known three dimensional structure supports the presence of a so-called beta-ladder domain in the C-terminal part of LtxA. LtxA rapidly killed K562 target cells transfected to express β(2) integrin. Cells expressing α(M)β(2) (CD11b/CD18) or α(X)β(2) (CD11c/CD18) were killed as efficiently as cells expressing α(L)β(2). Erythrocytes, which do not express β(2) integrins, were lysed more slowly. In ligand blotting experiments, LtxA bound only to the β(2) chain (CD18). These data support a previous suggestion that CD18 harbors the major binding site for LtxA as well as identifies integrins α(M)β(2) and α(X)β(2) as novel receptors for LtxA.
Collapse
|
46
|
Vorup-Jensen T. On the roles of polyvalent binding in immune recognition: perspectives in the nanoscience of immunology and the immune response to nanomedicines. Adv Drug Deliv Rev 2012; 64:1759-81. [PMID: 22705545 DOI: 10.1016/j.addr.2012.06.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2012] [Revised: 06/06/2012] [Accepted: 06/08/2012] [Indexed: 12/31/2022]
Abstract
Immunology often conveys the image of large molecules, either in the soluble state or in the membrane of leukocytes, forming multiple contacts with a target for actions of the immune system. Avidity names the ability of a polyvalent molecule to form multiple connections of the same kind with ligands tethered to the same surface. Polyvalent interactions are vastly stronger than their monovalent equivalent. In the present review, the functional consequences of polyvalent interactions are explored in a perspective of recent theoretical advances in understanding the thermodynamics of such binding. From insights on the structural biology of soluble pattern recognition molecules as well as adhesion molecules in the cell membranes or in their proteolytically shed form, this review documents the prominent role of polyvalent interactions in making the immune system a formidable barrier to microbial infection as well as constituting a significant challenge to the application of nanomedicines.
Collapse
|
47
|
Jalilian B, Einarsson HB, Vorup-Jensen T. Glatiramer acetate in treatment of multiple sclerosis: a toolbox of random co-polymers for targeting inflammatory mechanisms of both the innate and adaptive immune system? Int J Mol Sci 2012; 13:14579-605. [PMID: 23203082 PMCID: PMC3509598 DOI: 10.3390/ijms131114579] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 10/23/2012] [Accepted: 11/05/2012] [Indexed: 01/17/2023] Open
Abstract
Multiple sclerosis is a disease of the central nervous system, resulting in the demyelination of neurons, causing mild to severe symptoms. Several anti-inflammatory treatments now play a significant role in ameliorating the disease. Glatiramer acetate (GA) is a formulation of random polypeptide copolymers for the treatment of relapsing-remitting MS by limiting the frequency of attacks. While evidence suggests the influence of GA on inflammatory responses, the targeted molecular mechanisms remain poorly understood. Here, we review the multiple pharmacological modes-of-actions of glatiramer acetate in treatment of multiple sclerosis. We discuss in particular a newly discovered interaction between the leukocyte-expressed integrin α(M)β(2) (also called Mac-1, complement receptor 3, or CD11b/CD18) and perspectives on the GA co-polymers as an influence on the function of the innate immune system.
Collapse
Affiliation(s)
- Babak Jalilian
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, Building 1242, DK-8000, Aarhus C, Denmark; E-Mails: (B.J.); (H.B.E.)
| | - Halldór Bjarki Einarsson
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, Building 1242, DK-8000, Aarhus C, Denmark; E-Mails: (B.J.); (H.B.E.)
| | - Thomas Vorup-Jensen
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, Building 1242, DK-8000, Aarhus C, Denmark; E-Mails: (B.J.); (H.B.E.)
| |
Collapse
|
48
|
Altschuh D, Björkelund H, Strandgård J, Choulier L, Malmqvist M, Andersson K. Deciphering complex protein interaction kinetics using Interaction Map. Biochem Biophys Res Commun 2012; 428:74-9. [PMID: 23063847 DOI: 10.1016/j.bbrc.2012.10.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 10/02/2012] [Indexed: 10/27/2022]
Abstract
Cellular receptor systems are expected to present complex ligand interaction patterns that cannot be evaluated assuming a simple one ligand:one receptor interaction model. We have previously evaluated heterogeneous interactions using an alternative method to regression analysis, called Interaction Map (IM). IM decomposes a time-resolved binding curve into its separate components. By replacing the reductionistic, scalar kinetic association rate constant k(a) and dissociation rate constant k(d) with a two-dimensional distribution of k(a) and k(d), it is possible to display heterogeneous data as a map where each peak corresponds to one of the components that contribute to the cumulative binding curve. Here we challenge the Interaction Map approach by artificially generating heterogeneous data from two known interactions, on either LigandTracer or Surface Plasmon Resonance devices. We prove the ability of IM to accurately decompose these man-made heterogeneous binding curves composed of two different interactions. We conclude that the Interaction Map approach is well suited for the analysis of complex binding data and forecast that it has a potential to resolve previously uninterpretable data, in particular those generated in cell-based assays.
Collapse
Affiliation(s)
- Danièle Altschuh
- Biotechnologie et signalisation cellulaire, Université de Strasbourg, CNRS, Irebs-ESBS, Boulevard Sébastien Brant, 67412 Illkirch, France.
| | | | | | | | | | | |
Collapse
|
49
|
Chua GL, Tang XY, Patra AT, Tan SM, Bhattacharjya S. Structure and binding interface of the cytosolic tails of αXβ2 integrin. PLoS One 2012; 7:e41924. [PMID: 22844534 PMCID: PMC3406025 DOI: 10.1371/journal.pone.0041924] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 06/26/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Integrins are signal transducer proteins involved in a number of vital physiological processes including cell adhesion, proliferation and migration. Integrin molecules are hetero-dimers composed of two distinct subunits, α and β. In humans, 18 α and 8 β subunits are combined into 24 different integrin molecules. Each of the subunit comprises a large extracellular domain, a single pass transmembrane segment and a cytosolic tail (CT). The CTs of integrins are vital for bidirectional signal transduction and in maintaining the resting state of the receptors. A large number of intracellular proteins have been found to interact with the CTs of integrins linking integrins to the cytoskeleton. METHODOLOGY/PRINCIPAL FINDINGS In this work, we have investigated structure and interactions of CTs of the leukocyte specific integrin αXβ2. We determined the atomic resolution structure of a myristoylated CT of αX in perdeuterated dodecylphosphocholine (DPC) by NMR spectroscopy. Our results reveal that the 35-residue long CT of αX adopts an α-helical conformation for residues F4-N17 at the N-terminal region. The remaining residues located at the C-terminal segment of αX delineate a long loop of irregular conformations. A segment of the loop maintains packing interactions with the helical structure by an extended non-polar surface of the αX CT. Interactions between αX and β2 CTs are demonstrated by (15)N-(1)H HSQC NMR experiments. We find that residues constituting the polar face of the helical conformation of αX are involved in interactions with the N-terminal residues of β2 CT. A docked structure of the CT complex indicates that a network of polar and/or salt-bridge interactions may sustain the heteromeric interactions. CONCLUSIONS/SIGNIFICANCE The current study provides important insights into the conservation of interactions and structures among different CTs of integrins.
Collapse
Affiliation(s)
- Geok-Lin Chua
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Xiao-Yan Tang
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Alok Tanala Patra
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Suet-Mien Tan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
50
|
The leucocyte β2 (CD18) integrins: the structure, functional regulation and signalling properties. Biosci Rep 2012; 32:241-69. [PMID: 22458844 DOI: 10.1042/bsr20110101] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Leucocytes are highly motile cells. Their ability to migrate into tissues and organs is dependent on cell adhesion molecules. The integrins are a family of heterodimeric transmembrane cell adhesion molecules that are also signalling receptors. They are involved in many biological processes, including the development of metazoans, immunity, haemostasis, wound healing and cell survival, proliferation and differentiation. The leucocyte-restricted β2 integrins comprise four members, namely αLβ2, αMβ2, αXβ2 and αDβ2, which are required for a functional immune system. In this paper, the structure, functional regulation and signalling properties of these integrins are reviewed.
Collapse
|