1
|
Li Y, Xu X, Wang HJ, Chen YC, Chen Y, Chiu J, Li L, Wang L, Wang J, Tang Z, Ren L, Li H, Wang X, Jin S, Wu Y, Huang M, Ju LA, Fang C. Endoplasmic Reticulum Protein 72 Regulates Integrin Mac-1 Activity to Influence Neutrophil Recruitment. Arterioscler Thromb Vasc Biol 2024; 44:e82-e98. [PMID: 38205640 DOI: 10.1161/atvbaha.123.319771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Integrins mediate the adhesion, crawling, and migration of neutrophils during vascular inflammation. Thiol exchange is important in the regulation of integrin functions. ERp72 (endoplasmic reticulum-resident protein 72) is a member of the thiol isomerase family responsible for the catalysis of disulfide rearrangement. However, the role of ERp72 in the regulation of Mac-1 (integrin αMβ2) on neutrophils remains elusive. METHODS Intravital microscopy of the cremaster microcirculation was performed to determine in vivo neutrophil movement. Static adhesion, flow chamber, and flow cytometry were used to evaluate in vitro integrin functions. Confocal fluorescent microscopy and coimmunoprecipitation were utilized to characterize the interactions between ERp72 and Mac-1 on neutrophil surface. Cell-impermeable probes and mass spectrometry were used to label reactive thiols and identify target disulfide bonds during redox exchange. Biomembrane force probe was performed to quantitatively measure the binding affinity of Mac-1. A murine model of acute lung injury induced by lipopolysaccharide was utilized to evaluate neutrophil-associated vasculopathy. RESULTS ERp72-deficient neutrophils exhibited increased rolling but decreased adhesion/crawling on inflamed venules in vivo and defective static adhesion in vitro. The defect was due to defective activation of integrin Mac-1 but not LFA-1 (lymphocyte function-associated antigen-1) using blocking or epitope-specific antibodies. ERp72 interacted with Mac-1 in lipid rafts on neutrophil surface leading to the reduction of the C654-C711 disulfide bond in the αM subunit that is critical for Mac-1 activation. Recombinant ERp72, via its catalytic motifs, increased the binding affinity of Mac-1 with ICAM-1 (intercellular adhesion molecule-1) and rescued the defective adhesion of ERp72-deficient neutrophils both in vitro and in vivo. Deletion of ERp72 in the bone marrow inhibited neutrophil infiltration, ameliorated tissue damage, and increased survival during murine acute lung injury. CONCLUSIONS Extracellular ERp72 regulates integrin Mac-1 activity by catalyzing disulfide rearrangement on the αM subunit and may be a novel target for the treatment of neutrophil-associated vasculopathy.
Collapse
Affiliation(s)
- Yaofeng Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases (Y.L., X.X., L.L., L.W., C.F.), Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xulin Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases (Y.L., X.X., L.L., L.W., C.F.), Huazhong University of Science and Technology, Wuhan, Hubei, China
- Tongji-Rongcheng Center for Biomedicine (X.X., C.F.), Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Haoqing Jerry Wang
- School of Biomedical Engineering, Faculty of Engineering (H.J.W., Y.C.C., L.A.J.), The University of Sydney, New South Wales, Australia
- The University of Sydney Nano Institute Sydney Nanoscience Hub (H.J.W., Y.C.C., L.A.J.), The University of Sydney, New South Wales, Australia
| | - Yiyao Catherine Chen
- Institute of Pathology, Tongji Hospital, Tongji Medical College (Y.C.), Huazhong University of Science and Technology, Wuhan, Hubei, China
- School of Biomedical Engineering, Faculty of Engineering (H.J.W., Y.C.C., L.A.J.), The University of Sydney, New South Wales, Australia
| | - Yaobing Chen
- The University of Sydney Nano Institute Sydney Nanoscience Hub (H.J.W., Y.C.C., L.A.J.), The University of Sydney, New South Wales, Australia
| | - Joyce Chiu
- Centenary Institute (J.C.), The University of Sydney, New South Wales, Australia
| | - Li Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases (Y.L., X.X., L.L., L.W., C.F.), Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases (Y.L., X.X., L.L., L.W., C.F.), Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinyu Wang
- School of Stomatology, Tongji Medical Collage (J.W.), Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration of Hubei Province, Wuhan, China (J.W.)
| | - Zhaoming Tang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College (Z.T.), Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lehao Ren
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College (L.R.), Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongliang Li
- Laboratory of Chinese Herbal Pharmacology, Department of Pharmacy, Renmin Hospital of Wuhan University, China (H.L., X.W.)
- Biomedical Research Institute, School of Pharmaceutical Sciences and Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (H.L., X.W.)
| | - Xuanbin Wang
- Laboratory of Chinese Herbal Pharmacology, Department of Pharmacy, Renmin Hospital of Wuhan University, China (H.L., X.W.)
- Biomedical Research Institute, School of Pharmaceutical Sciences and Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (H.L., X.W.)
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College (S.J.), Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Wu
- Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, China (Y.W.)
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fujian, China (M.H.)
| | - Lining Arnold Ju
- School of Biomedical Engineering, Faculty of Engineering (H.J.W., Y.C.C., L.A.J.), The University of Sydney, New South Wales, Australia
- The University of Sydney Nano Institute Sydney Nanoscience Hub (H.J.W., Y.C.C., L.A.J.), The University of Sydney, New South Wales, Australia
| | - Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases (Y.L., X.X., L.L., L.W., C.F.), Huazhong University of Science and Technology, Wuhan, Hubei, China
- Tongji-Rongcheng Center for Biomedicine (X.X., C.F.), Huazhong University of Science and Technology, Wuhan, Hubei, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China (C.F.)
| |
Collapse
|
2
|
Su Y, Luo Z, Sun D, Yang B, Li Q. The Force-Dependent Mechanism of an Integrin α4β7-MAdCAM-1 Interaction. Int J Mol Sci 2023; 24:16062. [PMID: 38003252 PMCID: PMC10670920 DOI: 10.3390/ijms242216062] [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: 09/13/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
The interaction between integrin α4β7 and mucosal vascular addressin cell-adhesion molecule-1 (MAdCAM-1) facilitates the adhesion of circulating lymphocytes to the surface of high endothelial venules in inflammatory bowel diseases (IBDs). Lymphocyte adhesion is a multistep cascade involving the tethering, rolling, stable adhesion, crawling, and migration of cells, with integrin α4β7 being involved in rolling and stable adhesions. Targeting the integrin α4β7-MAdCAM-1 interaction may help decrease inflammation in IBDs. This interaction is regulated by force; however, the underlying mechanism remains unknown. Here, we investigate this mechanism using a parallel plate flow chamber and atomic force microscopy. The results reveal an initial increase in the lifetime of the integrin α4β7-MAdCAM-1 interaction followed by a decrease with an increasing force. This was manifested in a two-state curve regulated via a catch-bond-slip-bond conversion regardless of Ca2+ and/or Mg2+ availability. In contrast, the mean rolling velocity of cells initially decreased and then increased with the increasing force, indicating the flow-enhanced adhesion. Longer tether lifetimes of single bonds and lower rolling velocities mediated by multiple bonds were observed in the presence of Mg2+ rather than Ca2+. Similar results were obtained when examining the adhesion to substrates co-coated with chemokine CC motif ligand 25 and MAdCAM-1, as opposed to substrates coated with MAdCAM-1 alone. In conclusion, the integrin α4β7-MAdCAM-1 interaction occurs via ion- and cytokine-dependent flow-enhanced adhesion processes and is regulated via a catch-bond mechanism.
Collapse
Affiliation(s)
- Youmin Su
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China; (Y.S.); (Z.L.); (D.S.)
- Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, South China University of Technology, Guangzhou 510006, China
| | - Zhiqing Luo
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China; (Y.S.); (Z.L.); (D.S.)
| | - Dongshan Sun
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China; (Y.S.); (Z.L.); (D.S.)
| | - Bishan Yang
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China; (Y.S.); (Z.L.); (D.S.)
| | - Quhuan Li
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China; (Y.S.); (Z.L.); (D.S.)
- Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
3
|
Jenkins E, Körbel M, O'Brien-Ball C, McColl J, Chen KY, Kotowski M, Humphrey J, Lippert AH, Brouwer H, Santos AM, Lee SF, Davis SJ, Klenerman D. Antigen discrimination by T cells relies on size-constrained microvillar contact. Nat Commun 2023; 14:1611. [PMID: 36959206 PMCID: PMC10036606 DOI: 10.1038/s41467-023-36855-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 02/21/2023] [Indexed: 03/25/2023] Open
Abstract
T cells use finger-like protrusions called 'microvilli' to interrogate their targets, but why they do so is unknown. To form contacts, T cells must overcome the highly charged, barrier-like layer of large molecules forming a target cell's glycocalyx. Here, T cells are observed to use microvilli to breach a model glycocalyx barrier, forming numerous small (<0.5 μm diameter) contacts each of which is stabilized by the small adhesive protein CD2 expressed by the T cell, and excludes large proteins including CD45, allowing sensitive, antigen dependent TCR signaling. In the absence of the glycocalyx or when microvillar contact-size is increased by enhancing CD2 expression, strong signaling occurs that is no longer antigen dependent. Our observations suggest that, modulated by the opposing effects of the target cell glycocalyx and small adhesive proteins, the use of microvilli equips T cells with the ability to effect discriminatory receptor signaling.
Collapse
Affiliation(s)
- Edward Jenkins
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Markus Körbel
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Caitlin O'Brien-Ball
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - James McColl
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Kevin Y Chen
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Mateusz Kotowski
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Jane Humphrey
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Anna H Lippert
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Heather Brouwer
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Steven F Lee
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Simon J Davis
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK.
| |
Collapse
|
4
|
SSTR2 as an anatomical imaging marker and a safety switch to monitor and manage CAR T cell toxicity. Sci Rep 2022; 12:20932. [PMID: 36463361 PMCID: PMC9719480 DOI: 10.1038/s41598-022-25224-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/28/2022] [Indexed: 12/07/2022] Open
Abstract
The ability to image adoptively transferred T cells in the body and to eliminate them to avoid toxicity will be vital for chimeric antigen receptor (CAR) T cell therapy, particularly against solid tumors with higher risk of off-tumor toxicity. Previously, we have demonstrated the utility of somatostatin receptor 2 (SSTR2) for CAR T cell imaging, illustrating the expansion and contraction of CAR T cells in tumor as well as off-tumor expansion. Using intercellular adhesion molecule 1 (ICAM-1)-specific CAR T cells that secrete interleukin (IL)-12 as a model, herein we examined the potential of SSTR2 as a safety switch when combined with the SSTR2-specific maytansine-octreotate conjugate PEN-221. Constitutive secretion of IL-12 led to continuous expansion of CAR T cells after rapid elimination of tumors, causing systemic toxicity in mice with intact MHC expression. Treatment with PEN-221 rapidly reduced the abundance of CAR T cells, decreasing the severity of xenogeneic graft-versus-host disease (GvHD), and prolonged survival. Our study supports the development of SSTR2 as a single genetic marker for CAR T cells that is readily applicable to humans both for anatomical detection of T cell distribution and an image-guided safety switch for rapid elimination of CAR T cells.
Collapse
|
5
|
Humanized β2 Integrin-Expressing Hoxb8 Cells Serve as Model to Study Integrin Activation. Cells 2022; 11:cells11091532. [PMID: 35563841 PMCID: PMC9102476 DOI: 10.3390/cells11091532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/22/2022] [Accepted: 04/30/2022] [Indexed: 12/10/2022] Open
Abstract
The use of cell-based reporter systems has provided valuable insights into the molecular mechanisms of integrin activation. However, current models have significant drawbacks because their artificially expressed integrins cannot be regulated by either physiological stimuli or endogenous signaling pathways. Here, we report the generation of a Hoxb8 cell line expressing human β2 integrin that functionally replaced the deleted mouse ortholog. Hoxb8 cells are murine hematopoietic progenitor cells that can be efficiently differentiated into neutrophils and macrophages resembling their primary counterparts. Importantly, these cells can be stimulated by physiological stimuli triggering classical integrin inside-out signaling pathways, ultimately leading to β2 integrin conformational changes that can be recorded by the conformation-specific antibodies KIM127 and mAb24. Moreover, these cells can be efficiently manipulated via the CRISPR/Cas9 technique or retroviral vector systems. Deletion of the key integrin regulators talin1 and kindlin3 or expression of β2 integrins with mutations in their binding sites abolished both integrin extension and full activation regardless of whether only one or both activators no longer bind to the integrin. Moreover, humanized β2 integrin Hoxb8 cells represent a valuable new model for rapidly testing the role of putative integrin regulators in controlling β2 integrin activity in a physiological context.
Collapse
|
6
|
Mickolajczyk KJ, Olinares PDB, Chait BT, Liu S, Kapoor TM. The MIDAS domain of AAA mechanoenzyme Mdn1 forms catch bonds with two different substrates. eLife 2022; 11:73534. [PMID: 35147499 PMCID: PMC8837202 DOI: 10.7554/elife.73534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/19/2022] [Indexed: 12/21/2022] Open
Abstract
Catch bonds are a form of mechanoregulation wherein protein-ligand interactions are strengthened by the application of dissociative tension. Currently, the best-characterized examples of catch bonds are between single protein-ligand pairs. The essential AAA (ATPase associated with diverse cellular activities) mechanoenzyme Mdn1 drives at least two separate steps in ribosome biogenesis, using its MIDAS domain to extract the ubiquitin-like (UBL) domain-containing proteins Rsa4 and Ytm1 from ribosomal precursors. However, it must subsequently release these assembly factors to reinitiate the enzymatic cycle. The mechanism underlying the switching of the MIDAS-UBL interaction between strongly and weakly bound states is unknown. Here, we use optical tweezers to investigate the force dependence of MIDAS-UBL binding. Parallel experiments with Rsa4 and Ytm1 show that forces up to ~4 pN, matching the magnitude of force produced by AAA proteins similar to Mdn1, enhance the MIDAS domain binding lifetime up to 10-fold, and higher forces accelerate dissociation. Together, our studies indicate that Mdn1's MIDAS domain can form catch bonds with more than one UBL substrate, and provide insights into how mechanoregulation may contribute to the Mdn1 enzymatic cycle during ribosome biogenesis.
Collapse
Affiliation(s)
- Keith J Mickolajczyk
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, United States
| | - Paul Dominic B Olinares
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, United States
| | - Brian T Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, United States
| | - Shixin Liu
- Laboratory of Nanoscale Biophysics and Biochemistry, The Rockefeller University, New York, United States
| | - Tarun M Kapoor
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, United States
| |
Collapse
|
7
|
Al-Aghbar MA, Jainarayanan AK, Dustin ML, Roffler SR. The interplay between membrane topology and mechanical forces in regulating T cell receptor activity. Commun Biol 2022; 5:40. [PMID: 35017678 PMCID: PMC8752658 DOI: 10.1038/s42003-021-02995-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/21/2021] [Indexed: 12/20/2022] Open
Abstract
T cells are critically important for host defense against infections. T cell activation is specific because signal initiation requires T cell receptor (TCR) recognition of foreign antigen peptides presented by major histocompatibility complexes (pMHC) on antigen presenting cells (APCs). Recent advances reveal that the TCR acts as a mechanoreceptor, but it remains unclear how pMHC/TCR engagement generates mechanical forces that are converted to intracellular signals. Here we propose a TCR Bending Mechanosignal (TBM) model, in which local bending of the T cell membrane on the nanometer scale allows sustained contact of relatively small pMHC/TCR complexes interspersed among large surface receptors and adhesion molecules on the opposing surfaces of T cells and APCs. Localized T cell membrane bending is suggested to increase accessibility of TCR signaling domains to phosphorylation, facilitate selective recognition of agonists that form catch bonds, and reduce noise signals associated with slip bonds.
Collapse
Affiliation(s)
- Mohammad Ameen Al-Aghbar
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Department of Translational Medicine, Sidra Medicine, Doha, Qatar
| | - Ashwin K Jainarayanan
- Interdisciplinary Bioscience Doctoral Training Program and Exeter College, University of Oxford, Oxford, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
8
|
Khalaf NB, Al-Μehatab D, Fathallah DM. Vascular endothelial ERp72 is involved in the inflammatory response in a rat model of skeletal muscle injury. Mol Med Rep 2021; 23:186. [PMID: 33398381 PMCID: PMC7809907 DOI: 10.3892/mmr.2021.11825] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/25/2020] [Indexed: 11/05/2022] Open
Abstract
The vascular inflammatory response involves the coordinated action of a large network of molecular mediators and culminates in the transmigration of leukocytes into the site of inflammation. Inflammatory mediators include a variety of protein families, including adhesion molecules such as integrins and members of the immunoglobulin superfamily, as well as other cytokines and chemokines. In this study, a rat model of traumatic skeletal muscle injury was used to demonstrate endoplasmic reticulum resident protein 72 (ERp72) overexpression in the early phase of the inflammatory response that follows skeletal muscle injury. Reverse transcription‑quantitative PCR, western blotting, dual‑labeling immunohistochemistry and immunofluorescence experiments confirmed that ERp72 was expressed on the endothelial cells of blood vessels present at the injured area. In addition, a cell‑based neutrophil adhesion assay indicated that a polyclonal antibody specific for ERp72 significantly reduced adhesion of neutrophils to activated human umbilical vein endothelial cells (35% reduction). These data suggested that ERp72 expression on vascular endothelial cells may play a role in skeletal muscle inflammation and could be considered as a target for the modulation of leukocyte‑endothelial cell interactions in an inflammatory setting.
Collapse
Affiliation(s)
- Noureddine Ben Khalaf
- Department of Life Sciences, Health Biotechnology Program, College of Graduate Studies, King Fahd Chair for Health Biotechnology, Arabian Gulf University, Manama 329, Bahrain
| | - Dalal Al-Μehatab
- Department of Life Sciences, Health Biotechnology Program, College of Graduate Studies, King Fahd Chair for Health Biotechnology, Arabian Gulf University, Manama 329, Bahrain
| | - Dahmani M Fathallah
- Department of Life Sciences, Health Biotechnology Program, College of Graduate Studies, King Fahd Chair for Health Biotechnology, Arabian Gulf University, Manama 329, Bahrain
| |
Collapse
|
9
|
Ca 2+-based allosteric switches and shape shifting in RGLG1 VWA domain. Comput Struct Biotechnol J 2020; 18:821-833. [PMID: 32308929 PMCID: PMC7155146 DOI: 10.1016/j.csbj.2020.03.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022] Open
Abstract
RGLG1 is an E3 ubiquitin ligase in Arabidopsis thaliana that participates in ABA signaling and regulates apical dominance. Here, we present crystal structures of RGLG1 VWA domain, revealing two novel calcium ions binding sites (NCBS1 and NCBS2). Furthermore, the structures with guided mutagenesis in NCBS1 prove that Ca2+ ions play important roles in controlling conformational change of VWA, which is stabilized in open state with Ca2+ bound and converted to closed state after Ca2+ removal. This allosteric regulation mechanism is distinct from the ever reported one involving the C-terminal helix in integrin α and β I domains. The mutation of a key residue in NCBS2 do not abolish its Ca2+-binding potential, with no conformational change. MD simulations reveals that open state of RGLG1 VWA has higher ligand affinity than its closed state, consisting with integrin. Structural comparison of ion-free-MIDAS with Mg2+-MIDAS reveals that Mg2+ binding to MIDAS does not induce conformational change. With acquisition of first structure of plant VWA domain in both open state and closed state, we carefully analyze the conformational change and propose a totally new paradigm for its transition of open-closed states, which will be of great value for guiding future researches on VWA proteins and their important biological significance.
Collapse
|
10
|
Protein disulfide isomerase in cardiovascular disease. Exp Mol Med 2020; 52:390-399. [PMID: 32203104 PMCID: PMC7156431 DOI: 10.1038/s12276-020-0401-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/20/2020] [Accepted: 02/04/2020] [Indexed: 01/07/2023] Open
Abstract
Protein disulfide isomerase (PDI) participates in the pathogenesis of numerous diseases. Increasing evidence indicates that intravascular cell-derived PDI plays an important role in the initiation and progression of cardiovascular diseases, including thrombosis and vascular inflammation. Recent studies with PDI conditional knockout mice have advanced our understanding of the function of cell-specific PDI in disease processes. Furthermore, the identification and development of novel small-molecule PDI inhibitors has led into a new era of PDI research that transitioned from the bench to bedside. In this review, we will discuss recent findings on the regulatory role of PDI in cardiovascular disease. Efforts to untangle the functions of a large family of enzymes could lead researchers to new therapies for diverse cardiovascular diseases. Members of the protein disulfide isomerase (PDI) family chemically modify other proteins in ways that can alter both their structure and biological activity. Jaehyung Cho of the University of Illinois at Chicago, USA and coworkers have reviewed numerous studies linking PDI with cardiovascular diseases, including thrombosis, heart attack, vascular inflammation, and stroke. The authors also report progress in developing small-molecule PDI inhibitors that could yield the treatment for these conditions.
Collapse
|
11
|
MacKay L, Khadra A. The bioenergetics of integrin-based adhesion, from single molecule dynamics to stability of macromolecular complexes. Comput Struct Biotechnol J 2020; 18:393-416. [PMID: 32128069 PMCID: PMC7044673 DOI: 10.1016/j.csbj.2020.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/22/2022] Open
Abstract
The forces actively generated by motile cells must be transmitted to their environment in a spatiotemporally regulated manner, in order to produce directional cellular motion. This task is accomplished through integrin-based adhesions, large macromolecular complexes that link the actin-cytoskelton inside the cell to its external environment. Despite their relatively large size, adhesions exhibit rapid dynamics, switching between assembly and disassembly in response to chemical and mechanical cues exerted by cytoplasmic biochemical signals, and intracellular/extracellular forces, respectively. While in material science, force typically disrupts adhesive contact, in this biological system, force has a more nuanced effect, capable of causing assembly or disassembly. This initially puzzled experimentalists and theorists alike, but investigation into the mechanisms regulating adhesion dynamics have progressively elucidated the origin of these phenomena. This review provides an overview of recent studies focused on the theoretical understanding of adhesion assembly and disassembly as well as the experimental studies that motivated them. We first concentrate on the kinetics of integrin receptors, which exhibit a complex response to force, and then investigate how this response manifests itself in macromolecular adhesion complexes. We then turn our attention to studies of adhesion plaque dynamics that link integrins to the actin-cytoskeleton, and explain how force can influence the assembly/disassembly of these macromolecular structure. Subsequently, we analyze the effect of force on integrins populations across lengthscales larger than single adhesions. Finally, we cover some theoretical studies that have considered both integrins and the adhesion plaque and discuss some potential future avenues of research.
Collapse
Affiliation(s)
- Laurent MacKay
- Department of Physiology, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada
| | - Anmar Khadra
- Department of Physiology, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada
| |
Collapse
|
12
|
Haghayegh Jahromi N, Marchetti L, Moalli F, Duc D, Basso C, Tardent H, Kaba E, Deutsch U, Pot C, Sallusto F, Stein JV, Engelhardt B. Intercellular Adhesion Molecule-1 (ICAM-1) and ICAM-2 Differentially Contribute to Peripheral Activation and CNS Entry of Autoaggressive Th1 and Th17 Cells in Experimental Autoimmune Encephalomyelitis. Front Immunol 2020; 10:3056. [PMID: 31993059 PMCID: PMC6970977 DOI: 10.3389/fimmu.2019.03056] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/16/2019] [Indexed: 12/22/2022] Open
Abstract
In experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), myelin-specific T cells are activated in the periphery and differentiate in T helper (Th) 1 and Th17 effector cells, which cross the blood-brain barrier (BBB) to reach the central nervous system (CNS), where they induce neuroinflammation. Here, we explored the role of intercellular adhesion molecule-1 (ICAM-1) and ICAM-2 in the activation of naïve myelin-specific T cells and in the subsequent migration of differentiated encephalitogenic Th1 and Th17 cells across the BBB in vitro and in vivo. While on antigen-presenting cells ICAM-1, but not ICAM-2 was required for the activation of naïve CD4+ T cells, endothelial ICAM-1 and ICAM-2 mediated both Th1 and Th17 cell migration across the BBB. ICAM-1/-2-deficient mice developed ameliorated typical and atypical EAE transferred by encephalitogenic Th1 and Th17 cells, respectively. Our study underscores important yet cell-specific contributions for ICAM-1 and ICAM-2 in EAE pathogenesis.
Collapse
Affiliation(s)
| | - Luca Marchetti
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Federica Moalli
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Donovan Duc
- Laboratories of Neuroimmunology, Division of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, Epalinges, Switzerland
| | - Camilla Basso
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Heidi Tardent
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Elisa Kaba
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Caroline Pot
- Laboratories of Neuroimmunology, Division of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, Epalinges, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Department of Biology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Jens V Stein
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | |
Collapse
|
13
|
Exosomal regulation of lymphocyte homing to the gut. Blood Adv 2020; 3:1-11. [PMID: 30591532 DOI: 10.1182/bloodadvances.2018024877] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/28/2018] [Indexed: 12/12/2022] Open
Abstract
Exosomes secreted from T cells have been shown to affect dendritic cells, cancer cells, and other T cells. However, little is known about how T-cell exosomes (T exosomes) modulate endothelial cell functions in the context of tissue-specific homing. Here, we study the roles of T exosomes in the regulation of gut-specific T-cell homing. The gut-tropic T cells induced by retinoic acid secrete the exosomes that upregulate integrin α4β7 binding to the MAdCAM-1 expressed on high endothelial venules in the gut. T exosomes were preferentially distributed to the villi of the small intestine in an α4β7-dependent manner. Exosomes from gut-tropic T cells suppressed the expression of MAdCAM-1 in the small intestine, thereby inhibiting T-cell homing to the gut. Moreover, microRNA (miRNA) profiling analysis has shown that exosomes from gut-tropic T cells were enriched with miRNAs targeting NKX2.3, a transcription factor critical to MAdCAM-1 expression. Taken together, our study proposes that α4β7-expressing T exosomes distribute themselves to the small intestine and modify the expression of microenvironmental tissues such that any subsequent lymphocyte homing is precluded. This may represent a novel mechanism by which excessive lymphocyte homing to the intestinal tissues is downsized.
Collapse
|
14
|
Manufacturing and preclinical validation of CAR T cells targeting ICAM-1 for advanced thyroid cancer therapy. Sci Rep 2019; 9:10634. [PMID: 31337787 PMCID: PMC6650612 DOI: 10.1038/s41598-019-46938-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022] Open
Abstract
While the majority of thyroid cancer patients are easily treatable, those with anaplastic or poorly differentiated recurrent thyroid carcinomas have a very poor prognosis with a median survival of less than a year. Previously, we have shown a significant correlation between ICAM-1 overexpression and malignancy in thyroid cancer, and have pioneered the use of ICAM-1 targeted CAR T cells as a novel treatment modality. For clinical translation of this novel modality, we designed CAR T cells possessing micromolar rather than nanomolar affinity to ICAM-1 to avoid cytotoxicity in normal cells with basal levels of ICAM-1 expression. Herein, we report the automated process of CAR T cell manufacturing with CliniMACS Prodigy (Miltenyi Biotec) using cryopreserved peripheral blood leukocytes from apheresis collections. Using Prodigy, thawed leukopak cells were enriched for CD4+ and CD8+ T cells, subjected to double transduction using lentiviral vector, and expanded in culture for a total of 10 days with a final yield of 2–4 × 109 cells. The resulting CAR T cells were formulated for cryopreservation to be used directly for infusion into patients after thawing with no further processing. We examined cross-reactivity of CAR T cells toward both human and murine ICAM-1 and ICAM-1 expression in human and mouse tissues to demonstrate that both efficacy and on-target, off-tumor toxicity can be studied in our preclinical model. Selective anti-tumor activity in the absence of toxicity provides proof-of-concept that micromolar affinity tuned CAR T cells can be used to target tumors expressing high levels of antigen while avoiding normal tissues expressing basal levels of the same antigen. These studies support the initiation of a phase I study to evaluate the safety and potential efficacy of micromolar affinity tuned CAR T cells against newly diagnosed anaplastic and refractory or recurrent thyroid cancers.
Collapse
|
15
|
Walling BL, Kim M. LFA-1 in T Cell Migration and Differentiation. Front Immunol 2018; 9:952. [PMID: 29774029 PMCID: PMC5943560 DOI: 10.3389/fimmu.2018.00952] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/17/2018] [Indexed: 01/21/2023] Open
Abstract
Maintenance of homeostatic immune surveillance and development of effective adaptive immune responses require precise regulation of spatial and temporal lymphocyte trafficking throughout the body to ensure pathogen clearance and memory generation. Dysregulation of lymphocyte activation and migration can lead to impaired adaptive immunity, recurrent infections, and an array of autoimmune diseases and chronic inflammation. Central to the recruitment of T cells, integrins are cell surface receptors that regulate adhesion, signal transduction, and migration. With 24 integrin pairs having been discovered to date, integrins are defined not only by the composition of the heterodimeric pair but by cell-type specific expression and their ligands. Furthermore, integrins not only facilitate adhesion but also induce intracellular signaling and have recently been uncovered as mechanosensors providing additional complexity to the signaling pathways. Among several leukocyte-specific integrins, lymphocyte function-associated antigen-1 (LFA-1 or αLβ2; CD11a/CD18) is a key T cell integrin, which plays a major role in regulating T cell activation and migration. Adhesion to LFA-1's ligand, intracellular adhesion receptor 1 (ICAM-1) facilitates firm endothelium adhesion, prolonged contact with antigen-presenting cells, and binding to target cells for killing. While the downstream signaling pathways utilized by LFA-1 are vastly conserved they allow for highly disparate responses. Here, we summarize the roles of LFA-1 and ongoing studies to better understand its functions and regulation.
Collapse
Affiliation(s)
- Brandon L Walling
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
16
|
Swaim CD, Scott AF, Canadeo LA, Huibregtse JM. Extracellular ISG15 Signals Cytokine Secretion through the LFA-1 Integrin Receptor. Mol Cell 2017; 68:581-590.e5. [PMID: 29100055 DOI: 10.1016/j.molcel.2017.10.003] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/31/2017] [Accepted: 10/03/2017] [Indexed: 12/30/2022]
Abstract
ISG15 is a ubiquitin-like protein that functions in innate immunity both as an intracellular protein modifier and as an extracellular signaling molecule that stimulates IFN-γ secretion. The extracellular function, important for resistance to mycobacterial disease, has remained biochemically uncharacterized. We have established an NK-92 cell-based assay for IFN-γ release, identified residues critical for ISG15 signaling, and identified the cell surface receptor as LFA-1 (CD11a/CD18; αLβ2 integrin). LFA-1 inhibition blocked IFN-γ secretion, splenocytes from CD11a-/- mice did not respond to ISG15, and ISG15 bound directly to the αI domain of CD11a in vitro. ISG15 also enhanced secretion of IL-10, indicating a broader role for ISG15 in cytokine signaling. ISG15 engagement of LFA-1 led to the activation of SRC family kinases (SFKs) and SFK inhibition blocked cytokine secretion. These findings establish the molecular basis of the extracellular function of ISG15 and the initial outside-in signaling events that drive ISG15-dependent cytokine secretion.
Collapse
Affiliation(s)
- Caleb D Swaim
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Ariella F Scott
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Larissa A Canadeo
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Jon M Huibregtse
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
17
|
Zhang X, Li L, Li N, Shu X, Zhou L, Lü S, Chen S, Mao D, Long M. Salt bridge interactions within the β 2 integrin α 7 helix mediate force-induced binding and shear resistance ability. FEBS J 2017; 285:261-274. [PMID: 29150976 DOI: 10.1111/febs.14335] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/23/2017] [Accepted: 11/14/2017] [Indexed: 01/13/2023]
Abstract
The functional performance of the αI domain α7 helix in β2 integrin activation depends on the allostery of the α7 helix, which axially slides down; therefore, it is critical to elucidate what factors regulate the allostery. In this study, we determined that there were two conservative salt bridge interaction pairs that constrain both the upper and bottom ends of the α7 helix. Molecular dynamics (MD) simulations for three β2 integrin members, lymphocyte function-associated antigen-1 (LFA-1; αL β2 ), macrophage-1 antigen (Mac-1; αM β2 ) and αx β2 , indicated that the magnitude of the salt bridge interaction is related to the stability of the αI domain and the strength of the corresponding force-induced allostery. The disruption of the salt bridge interaction, especially with double mutations in both salt bridges, significantly reduced the force-induced allostery time for all three members. The effects of salt bridge interactions of the αI domain α7 helix on β2 integrin conformational stability and allostery were experimentally validated using Mac-1 constructs. The results demonstrated that salt bridge mutations did not alter the conformational state of Mac-1, but they did increase the force-induced ligand binding and shear resistance ability, which was consistent with MD simulations. This study offers new insight into the importance of salt bridge interaction constraints of the αI domain α7 helix and external force for β2 integrin function.
Collapse
Affiliation(s)
- Xiao Zhang
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Linda Li
- College of Bioengineering, Chongqing University, China
| | - Ning Li
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Xinyu Shu
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Lüwen Zhou
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Shouqin Lü
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Shenbao Chen
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Debin Mao
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Mian Long
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
18
|
Micromolar affinity CAR T cells to ICAM-1 achieves rapid tumor elimination while avoiding systemic toxicity. Sci Rep 2017; 7:14366. [PMID: 29085043 PMCID: PMC5662687 DOI: 10.1038/s41598-017-14749-3] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/17/2017] [Indexed: 01/04/2023] Open
Abstract
Adoptive transfer of high-affinity chimeric antigen receptor (CAR) T cells targeting hematological cancers has yielded impressive clinical results. However, safety concerns regarding target expression on healthy tissue and poor efficacy have hampered application to solid tumors. Here, a panel of affinity-variant CARs were constructed targeting overexpressed ICAM-1, a broad tumor biomarker, using its physiological ligand, LFA-1. Anti-tumor T cell potency in vitro was directly proportional to CAR affinity and ICAM-1 density. In a solid tumor mouse model allowing simultaneous monitoring of anti-tumor potency and systemic off-tumor toxicity, micromolar affinity CAR T cells demonstrated superior anti-tumor efficacy and safety compared to their nanomolar counterparts. Longitudinal T cell tracking by PET/CT and concurrent cytokine measurement revealed superior expansion and contraction kinetics of micromolar affinity CAR T cells. Therefore, we developed an ICAM-1 specific CAR with broad anti-tumor applicability that utilized a reduced affinity targeting strategy to significantly boost efficacy and safety.
Collapse
|
19
|
Abdullahi M, Olotu FA, Soliman ME. Dynamics of allosteric modulation of lymphocyte function associated antigen-1 closure-open switch: unveiling the structural mechanisms associated with outside-in signaling activation. Biotechnol Lett 2017; 39:1843-1851. [PMID: 28918491 DOI: 10.1007/s10529-017-2432-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 08/31/2017] [Indexed: 10/18/2022]
Abstract
OBJECTIVES To provide insight into the dynamics of the shape-shifting mechanistic events associated with the opening (activation) of Lymphocyte Function Associated Antigen-1 upon allosteric modulation by an activator, ICAM Binding Enhancer-667 (IBE-667), using molecular dynamics simulation. RESULTS Various parameters were used to appropriately describe and understand the sequence of events that characterized its activation across the simulation period such as residual distances, TriCα angles; as well as the dihedral angle. Our findings revealed a significant residual fluctuation and stability difference between both systems. Also, there was a synergistic coordination of the active MIDAS site by the downward pull of the α7 helix upon ligand binding, which appeared to be directly proportional to each other. CONCLUSION Allosteric binding of IBE-667, activated LFA-1 integrin as evidenced by residual motion at the MIDAS region which appears to be synergistically coordinated by the downward pull of the α7 helix.
Collapse
Affiliation(s)
- Maryam Abdullahi
- Molecular Modeling and Drug Design Research, Group School of Health Sciences, University of KwaZulu- Natal, Westville Campus, Durban, 4001, South Africa
| | - Fisayo A Olotu
- Molecular Modeling and Drug Design Research, Group School of Health Sciences, University of KwaZulu- Natal, Westville Campus, Durban, 4001, South Africa
| | - Mahmoud E Soliman
- Molecular Modeling and Drug Design Research, Group School of Health Sciences, University of KwaZulu- Natal, Westville Campus, Durban, 4001, South Africa. .,College of Pharmacy and Pharmaceutical Sciences, Florida Agricultural and Mechanical University (FAMU), Tallahassee, FL, 32307, USA. .,Department of Pharmaceutical Organic Chemistry Faculty of Pharmacy, Zagazig University, Zagazig, Egypt.
| |
Collapse
|
20
|
Dramatic and concerted conformational changes enable rhodocetin to block α2β1 integrin selectively. PLoS Biol 2017; 15:e2001492. [PMID: 28704364 PMCID: PMC5509089 DOI: 10.1371/journal.pbio.2001492] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 06/15/2017] [Indexed: 01/08/2023] Open
Abstract
The collagen binding integrin α2β1 plays a crucial role in hemostasis, fibrosis, and cancer progression amongst others. It is specifically inhibited by rhodocetin (RC), a C-type lectin-related protein (CLRP) found in Malayan pit viper (Calloselasma rhodostoma) venom. The structure of RC alone reveals a heterotetramer arranged as an αβ and γδ subunit in a cruciform shape. RC specifically binds to the collagen binding A-domain of the integrin α2 subunit, thereby blocking collagen-induced platelet aggregation. However, until now, the molecular basis for this interaction has remained unclear. Here, we present the molecular structure of the RCγδ-α2A complex solved to 3.0 Å resolution. Our findings show that RC undergoes a dramatic structural reorganization upon binding to α2β1 integrin. Besides the release of the nonbinding RCαβ tandem, the RCγ subunit interacts with loop 2 of the α2A domain as result of a dramatic conformational change. The RCδ subunit contacts the integrin α2A domain in the “closed” conformation through its helix C. Combined with epitope-mapped antibodies, conformationally locked α2A domain mutants, point mutations within the α2A loop 2, and chemical modifications of the purified toxin protein, this molecular structure of RCγδ-α2A complex explains the inhibitory mechanism and specificity of RC for α2β1 integrin. In animals, collagen-mediated platelet aggregation is an essential component of the blood’s clotting response following vascular injury. A small group of snake venom toxins belonging to the C-type lectin protein family exert their harmful effects by directly targeting this pathway. Rhodocetin (RC) is a heterotetrameric protein found in the venom of the Malayan pit viper (C. rhodostoma). RC specifically binds α2β1 integrin, the key protein required for collagen-mediated platelet aggregation. In this study, we describe the interaction between RC and α2β1 integrin at atomic resolution. This study reveals that RC undergoes a massive structural reorganization upon α2β1 integrin binding, such that RC’s αβ subunit is released from its γδ subunit and a γδ-α2β1 integrin complex is formed. The inhibitory nature of this complex can be readily explained as RC binding along the top surface of the α2β1 integrin and directly above the collagen binding site. As a result, access of collagen to its binding site is blocked, thereby preventing collagen-mediated platelet aggregation.
Collapse
|
21
|
Li K, Cheng X, Tilevik A, Davis SJ, Zhu C. In situ and in silico kinetic analyses of programmed cell death-1 (PD-1) receptor, programmed cell death ligands, and B7-1 protein interaction network. J Biol Chem 2017; 292:6799-6809. [PMID: 28270509 DOI: 10.1074/jbc.m116.763888] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 03/06/2017] [Indexed: 12/13/2022] Open
Abstract
Programmed cell death-1 (PD-1) is an inhibitory receptor with an essential role in maintaining peripheral tolerance and is among the most promising immunotherapeutic targets for treating cancer, autoimmunity, and infectious diseases. A complete understanding of the consequences of PD-1 engagement by its ligands, PD-L1 and PD-L2, and of PD-L1 binding to B7-1 requires quantitative analysis of their interactions at the cell surface. We present here the first complete in situ kinetic analysis of the PD-1/PD-ligands/B7-1 system. Consistent with previous solution measurements, we observed higher in situ affinities for human (h) than murine (m) PD-1 interactions, stronger binding of hPD-1 to hPD-L2 than hPD-L1, and comparable binding of mPD-1 to both ligands. However, in contrast to the relatively weak solution affinities, the in situ affinities of PD-1 are as high as those of the T cell receptor for agonist pMHC and of LFA-1 (lymphocyte function-associated antigen 1) for ICAM-1 (intercellular adhesion molecule 1) but significantly lower than that of the B7-1/CTLA-4 interaction, suggesting a distinct basis for PD-1- versus CTLA-4-mediated inhibition. Notably, the in situ interactions of PD-1 are much stronger than that of B7-1 with PD-L1. Overall, the in situ affinity ranking greatly depends on the on-rate instead of the off-rate. In silico simulations predict that PD-1/PD-L1 interactions dominate at interfaces between activated T cells and mature dendritic cells and that these interactions will be highly sensitive to the dynamics of PD-L1 and PD-L2 expression. Our results provide a kinetic framework for better understanding inhibitory PD-1 activity in health and disease.
Collapse
Affiliation(s)
- Kaitao Li
- From the Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta 30332-0535, Georgia
| | - Xiaoxiao Cheng
- the Radcliffe Department of Medicine and Medical Research Council Human Immunology Unit, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DU, United Kingdom
| | - Andreas Tilevik
- the Systems Biology Research Centre, School of Bioscience, University of Skövde, Box 408, Skövde, Sweden, and
| | - Simon J Davis
- the Radcliffe Department of Medicine and Medical Research Council Human Immunology Unit, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DU, United Kingdom
| | - Cheng Zhu
- the Coulter Department of Biomedical Engineering, the Woodruff School of Mechanical Engineering, and the Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta 30332-0535, Georgia
| |
Collapse
|
22
|
Bazylewski P, Divigalpitiya R, Fanchini G. In situ Raman spectroscopy distinguishes between reversible and irreversible thiol modifications in l-cysteine. RSC Adv 2017. [DOI: 10.1039/c6ra25879d] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Reversibility of disulphide formation and breakage on l-cysteine examined through vibrational modes using in situ Raman spectroscopy.
Collapse
Affiliation(s)
- Paul Bazylewski
- Department of Physics and Astronomy
- University of Western Ontario
- London
- Canada
| | | | - Giovanni Fanchini
- Department of Physics and Astronomy
- University of Western Ontario
- London
- Canada
- Department of Chemistry
| |
Collapse
|
23
|
Fan Z, Ley K. Leukocyte arrest: Biomechanics and molecular mechanisms of β2 integrin activation. Biorheology 2016; 52:353-77. [PMID: 26684674 DOI: 10.3233/bir-15085] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Integrins are a group of heterodimeric transmembrane receptors that play essential roles in cell-cell and cell-matrix interaction. Integrins are important in many physiological processes and diseases. Integrins acquire affinity to their ligand by undergoing molecular conformational changes called activation. Here we review the molecular biomechanics during conformational changes of integrins, integrin functions in leukocyte biorheology (adhesive functions during rolling and arrest) and molecules involved in integrin activation.
Collapse
Affiliation(s)
- Zhichao Fan
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.,Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
24
|
Complement MASP-1 enhances adhesion between endothelial cells and neutrophils by up-regulating E-selectin expression. Mol Immunol 2016; 75:38-47. [PMID: 27219453 DOI: 10.1016/j.molimm.2016.05.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/05/2016] [Accepted: 05/07/2016] [Indexed: 12/31/2022]
Abstract
The complement system and neutrophil granulocytes are indispensable in the immune response against extracellular pathogens such as bacteria and fungi. Endothelial cells also participate in antimicrobial immunity largely by regulating the homing of leukocytes through their cytokine production and their pattern of cell surface adhesion molecules. We have previously shown that mannan-binding lectin-associated serine protease-1 (MASP-1), a complement lectin pathway enzyme, is able to activate endothelial cells by cleaving protease activated receptors, which leads to cytokine production and enables neutrophil chemotaxis. Therefore, we aimed to investigate how recombinant MASP-1 (rMASP-1) can modify the pattern of P-selectin, E-selectin, ICAM-1, ICAM-2, and VCAM-1 adhesion molecules in human umbilical vein endothelial cells (HUVEC), and whether these changes can enhance the adherence between endothelial cells and neutrophil granulocyte model cells (differentiated PLB-985). We found that HUVECs activated by rMASP-1 decreased the expression of ICAM-2 and increased that of E-selectin, whereas ICAM-1, VCAM-1 and P-selectin expression remained unchanged. Furthermore, these changes resulted in increased adherence between differentiated PLB-985 cells and endothelial cells. Our finding suggests that complement MASP-1 can increase adhesion between neutrophils and endothelial cells in a direct fashion. This is in agreement with our previous finding that MASP-1 increases the production of pro-inflammatory cytokines (such as IL-6 and IL-8) and chemotaxis, and may thereby boost neutrophil functions. This newly described cooperation between complement lectin pathway and neutrophils via endothelial cells may be an effective tool to enhance the antimicrobial immune response.
Collapse
|
25
|
Wald T, Osickova A, Masin J, Liskova PM, Petry-Podgorska I, Matousek T, Sebo P, Osicka R. Transmembrane segments of complement receptor 3 do not participate in cytotoxic activities but determine receptor structure required for action of Bordetella adenylate cyclase toxin. Pathog Dis 2016; 74:ftw008. [PMID: 26802078 DOI: 10.1093/femspd/ftw008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2016] [Indexed: 11/13/2022] Open
Abstract
Adenylate cyclase toxin-hemolysin (CyaA, ACT or AC-Hly) of the whooping cough agent Bordetella pertussis penetrates phagocytes expressing the integrin complement receptor 3 (CR3, CD11b/CD18, α(M)β(2) or Mac-1). CyaA translocates its adenylate cyclase (AC) enzyme domain into cell cytosol and catalyzes unregulated conversion of ATP to cAMP, thereby subverting cellular signaling. In parallel, CyaA forms small cation-selective membrane pores that permeabilize cells for potassium efflux, contributing to cytotoxicity of CyaA and eventually provoking colloid-osmotic cell lysis. To investigate whether the single-pass α-helical transmembrane segments of CR3 subunits CD11b and CD18 do directly participate in AC domain translocation and/or pore formation by the toxin, we expressed in CHO cells variants of CR3 that contained artificial transmembrane segments, or lacked the transmembrane segment(s) at all. The results demonstrate that the transmembrane segments of CR3 are not directly involved in the cytotoxic activities of CyaA but serve for maintaining CR3 in a conformation that is required for efficient toxin binding and action.
Collapse
Affiliation(s)
- Tomas Wald
- Institute of Microbiology of the CAS, v. v. i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Adriana Osickova
- Institute of Microbiology of the CAS, v. v. i., Videnska 1083, 142 20, Prague 4, Czech Republic Department of Biochemistry, Faculty of Science, Charles University in Prague, Hlavova 8, 128 43, Prague 2, Czech Republic
| | - Jiri Masin
- Institute of Microbiology of the CAS, v. v. i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Petra M Liskova
- Institute of Microbiology of the CAS, v. v. i., Videnska 1083, 142 20, Prague 4, Czech Republic Department of Genetics and Microbiology, Faculty of Science, Charles University in Prague, Vinicna 5, 128 44 Prague 2, Czech Republic
| | - Inga Petry-Podgorska
- Institute of Analytical Chemistry of the CAS, v. v. i., Veveri 97, 602 00 Brno, Czech Republic
| | - Tomas Matousek
- Institute of Analytical Chemistry of the CAS, v. v. i., Veveri 97, 602 00 Brno, Czech Republic
| | - Peter Sebo
- Institute of Microbiology of the CAS, v. v. i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Radim Osicka
- Institute of Microbiology of the CAS, v. v. i., Videnska 1083, 142 20, Prague 4, Czech Republic
| |
Collapse
|
26
|
Small Molecule Targeting of Protein-Protein Interactions through Allosteric Modulation of Dynamics. Molecules 2015; 20:16435-45. [PMID: 26378508 PMCID: PMC6332300 DOI: 10.3390/molecules200916435] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/06/2015] [Accepted: 09/08/2015] [Indexed: 11/16/2022] Open
Abstract
The protein–protein interaction (PPI) target class is particularly challenging, but offers potential for “first in class” therapies. Most known PPI small molecules are orthosteric inhibitors but many PPI sites may be fundamentally intractable to this approach. One potential alternative is to consider more attractive, remote small molecule pockets; however, on the whole, allostery is poorly understood and difficult to discover and develop. Here we review the literature in order to understand the basis for allostery, especially as it can apply to PPIs. We suggest that the upfront generation of sophisticated and experimentally validated dynamic models of target proteins can aid in target choice and strategy for allosteric intervention to produce the required functional effect.
Collapse
|
27
|
Ishibashi M, Miyanaga Y, Matsuoka S, Kozuka J, Togashi Y, Kinashi T, Ueda M. Integrin LFA-1 regulates cell adhesion via transient clutch formation. Biochem Biophys Res Commun 2015; 464:459-66. [DOI: 10.1016/j.bbrc.2015.06.155] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 06/24/2015] [Indexed: 10/23/2022]
|
28
|
Park EJ, Yuki Y, Kiyono H, Shimaoka M. Structural basis of blocking integrin activation and deactivation for anti-inflammation. J Biomed Sci 2015; 22:51. [PMID: 26152212 PMCID: PMC4495637 DOI: 10.1186/s12929-015-0159-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/22/2015] [Indexed: 12/30/2022] Open
Abstract
Integrins mediate leukocyte accumulation to the sites of inflammation, thereby enhancing their potential as an important therapeutic target for inflammatory disorders. Integrin activation triggered by inflammatory mediators or signaling pathway is a key step to initiate leukocyte migration to inflamed tissues; however, an appropriately regulated integrin deactivation is indispensable for maintaining productive leukocyte migration. While typical integrin antagonists that block integrin activation target the initiation of leukocyte migration, a novel class of experimental compounds has been designed to block integrin deactivation, thereby perturbing the progression of cell migration. Current review discusses the mechanisms by which integrin is activated and subsequently deactivated by focusing on its structure-function relationship.
Collapse
Affiliation(s)
- Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Mie, 514-8507, Japan. .,Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.
| | - Yoshikazu Yuki
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan. .,International Research and Development Center for Mucosal Vaccine, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Mie, 514-8507, Japan.
| |
Collapse
|
29
|
Kukic P, Alvin Leung HT, Bemporad F, Aprile FA, Kumita JR, De Simone A, Camilloni C, Vendruscolo M. Structure and dynamics of the integrin LFA-1 I-domain in the inactive state underlie its inside-out/outside-in signaling and allosteric mechanisms. Structure 2015; 23:745-53. [PMID: 25773142 PMCID: PMC4396694 DOI: 10.1016/j.str.2014.12.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/04/2014] [Accepted: 12/06/2014] [Indexed: 01/13/2023]
Abstract
Lymphocyte function-associated antigen 1 (LFA-1) is an integrin that transmits information in two directions across the plasma membrane of leukocytes, in so-called outside-in and inside-out signaling mechanisms. To investigate the structural basis of these mechanisms, we studied the conformational space of the apo I-domain using replica-averaged metadynamics simulations in combination with nuclear magnetic resonance chemical shifts. We thus obtained a free energy landscape that reveals the existence of three conformational substates of this domain. The three substates include conformations similar to existing crystallographic structures of the low-affinity I-domain, the inactive I-domain with an allosteric antagonist inhibitor bound underneath α helix 7, and an intermediate affinity state of the I-domain. The multiple substates were validated with residual dipolar coupling measurements. These results suggest that the presence of three substates in the apo I-domain enables the precise regulation of the binding process that is essential for the physiological function of LFA-1.
Collapse
Affiliation(s)
- Predrag Kukic
- Department of Chemistry, Lensfield Road, University of Cambridge, Cambridge CB2 1EW, UK
| | - Hoi Tik Alvin Leung
- Department of Chemistry, Lensfield Road, University of Cambridge, Cambridge CB2 1EW, UK; Biozentrum, University of Basel, Klingelbergstrasse 50/70, Basel 4056, Switzerland
| | - Francesco Bemporad
- Department of Chemistry, Lensfield Road, University of Cambridge, Cambridge CB2 1EW, UK; Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università degli Studi di Firenze, Viale G. B. Morgagni 50, 50134, Firenze, Italy
| | - Francesco A Aprile
- Department of Chemistry, Lensfield Road, University of Cambridge, Cambridge CB2 1EW, UK
| | - Janet R Kumita
- Department of Chemistry, Lensfield Road, University of Cambridge, Cambridge CB2 1EW, UK
| | - Alfonso De Simone
- Division of Molecular Biosciences, Imperial College London, London SW7 2AZ, UK
| | - Carlo Camilloni
- Department of Chemistry, Lensfield Road, University of Cambridge, Cambridge CB2 1EW, UK
| | - Michele Vendruscolo
- Department of Chemistry, Lensfield Road, University of Cambridge, Cambridge CB2 1EW, UK.
| |
Collapse
|
30
|
Fu X, Xu Y, Wu C, Moy VT, Zhang XF. Anchorage-dependent binding of integrin I-domain to adhesion ligands. J Mol Recognit 2015; 28:385-92. [PMID: 25707989 DOI: 10.1002/jmr.2453] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/28/2014] [Accepted: 12/01/2014] [Indexed: 01/17/2023]
Abstract
The dynamic interactions between leukocyte integrin receptors and ligands in the vascular endothelium, extracellular matrix, or invading pathogens result in leukocyte adhesion, extravasation, and phagocytosis. This work examined the mechanical strength of the connection between iC3b, a complement component that stimulates phagocytosis, and the ligand-binding domain, the I-domain, of integrin αMβ2. Single-molecule force measurements of αM I-domain-iC3b complexes were conducted by atomic force microscope. Strikingly, depending on loading rates, immobilization of the I-domain via its C-terminus resulted in a 1.3-fold to 1.5-fold increase in unbinding force compared with I-domains immobilized via the N-terminus. The force spectra (unbinding force versus loading rate) of the I-domain-iC3b complexes revealed that the enhanced mechanical strength is due to a 2.4-fold increase in the lifetime of the I-domain-iC3b bond. Given the structural and functional similarity of all integrin I-domains, our result supports the existing allosteric regulatory model by which the ligand binding strength of integrin can be increased rapidly when a force is allowed to stretch the C-terminus of the I-domain. This type of mechanism may account for the rapid ligand affinity adjustment during leukocyte migration.
Collapse
Affiliation(s)
- Xin Fu
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | |
Collapse
|
31
|
Leung HTA, Kukic P, Camilloni C, Bemporad F, De Simone A, Aprile FA, Kumita JR, Vendruscolo M. NMR characterization of the conformational fluctuations of the human lymphocyte function-associated antigen-1 I-domain. Protein Sci 2014; 23:1596-606. [PMID: 25147050 DOI: 10.1002/pro.2538] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 08/13/2014] [Accepted: 08/19/2014] [Indexed: 01/13/2023]
Abstract
Lymphocyte function-associated antigen-1 (LFA-1) is an integrin protein that transmits information across the plasma membrane through the so-called inside-out and outside-in signaling mechanisms. To investigate these mechanisms, we carried out an NMR analysis of the dynamics of the LFA-1 I-domain, which has enabled us to characterize the motions of this domain on a broad range of timescales. We studied first the internal motions on the nanosecond timescale by spin relaxation measurements and model-free analysis. We then extended this analysis to the millisecond timescale motions by measuring (15) N-(1) H residual dipolar couplings of the backbone amide groups. We analyzed these results in the context of the three major conformational states of the I-domain using their corresponding X-ray crystallographic structures. Our results highlight the importance of the low-frequency motions of the LFA-1 I-domain in the inactive apo-state. We found in particular that α-helix 7 is in a position in the apo-closed state that cannot be fully described by any of the existing X-ray structures, as it appears to be in dynamic exchange between different conformations. This type of motion seems to represent an inherent property of the LFA-1 I-domain and might be relevant for controlling the access to the allosteric binding pocket, as well as for the downward displacement of α-helix 7 that is required for the activation of LFA-1.
Collapse
Affiliation(s)
- Hoi Tik Alvin Leung
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom; Biozentrum, University of Basel, Basel, CH-4056, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Chakraborty S, Núñez D, Hu SY, Domingo MP, Pardo J, Karmenyan A, Chiou A. FRET based quantification and screening technology platform for the interactions of leukocyte function-associated antigen-1 (LFA-1) with intercellular adhesion molecule-1 (ICAM-1). PLoS One 2014; 9:e102572. [PMID: 25032811 PMCID: PMC4102529 DOI: 10.1371/journal.pone.0102572] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 06/19/2014] [Indexed: 11/29/2022] Open
Abstract
The interaction between leukocyte function-associated antigen-1(LFA-1) and intercellular adhesion molecule-1 (ICAM-1) plays a pivotal role in cellular adhesion including the extravasation and inflammatory response of leukocytes, and also in the formation of immunological synapse. However, irregular expressions of LFA-1 or ICAM-1 or both may lead to autoimmune diseases, metastasis cancer, etc. Thus, the LFA-1/ICAM-1 interaction may serve as a potential therapeutic target for the treatment of these diseases. Here, we developed one simple 'in solution' steady state fluorescence resonance energy transfer (FRET) technique to obtain the dissociation constant (Kd) of the interaction between LFA-1 and ICAM-1. Moreover, we developed the assay into a screening platform to identify peptides and small molecules that inhibit the LFA-1/ICAM-1 interaction. For the FRET pair, we used Alexa Fluor 488-LFA-1 conjugate as donor and Alexa Fluor 555-human recombinant ICAM-1 (D1-D2-Fc) as acceptor. From our quantitative FRET analysis, the Kd between LFA-1 and D1-D2-Fc was determined to be 17.93±1.34 nM. Both the Kd determination and screening assay were performed in a 96-well plate platform, providing the opportunity to develop it into a high-throughput assay. This is the first reported work which applies FRET based technique to determine Kd as well as classifying inhibitors of the LFA-1/ICAM-1 interaction.
Collapse
Affiliation(s)
| | - David Núñez
- Instituto de Carboquímica, CSIC, Zaragoza, Spain
- Immune Effector Cells Group, Aragón Health Research Institute, Biomedical Research Centre of Aragón, Zaragoza, Spain
| | - Shih-Yang Hu
- Institute of Biophotonics, National Yang-Ming University, Taipei, Taiwan
| | - María Pilar Domingo
- Instituto de Carboquímica, CSIC, Zaragoza, Spain
- Immune Effector Cells Group, Aragón Health Research Institute, Biomedical Research Centre of Aragón, Zaragoza, Spain
| | - Julian Pardo
- Immune Effector Cells Group, Aragón Health Research Institute, Biomedical Research Centre of Aragón, Zaragoza, Spain
- Department of Biochemistry and Molecular and Cell Biology, Facultad de Ciencias, University of Zaragoza, Zaragoza, Spain
- Aragón I+D Foundation, Government of Aragon, Zaragoza, Spain
- Nanoscience Institute of Aragón, Aragón I+D Foundation, University of Zaragoza, Zaragoza, Spain
| | - Artashes Karmenyan
- Biophotonics & Molecular Imaging Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Eva Ma Gálvez
- Instituto de Carboquímica, CSIC, Zaragoza, Spain
- Immune Effector Cells Group, Aragón Health Research Institute, Biomedical Research Centre of Aragón, Zaragoza, Spain
| | - Arthur Chiou
- Institute of Biophotonics, National Yang-Ming University, Taipei, Taiwan
- Biophotonics & Molecular Imaging Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
33
|
Samadzadeh KM, Chun KC, Nguyen AT, Baker PM, Bains S, Lee ES. Monocyte activity is linked with abdominal aortic aneurysm diameter. J Surg Res 2014; 190:328-34. [PMID: 24726061 DOI: 10.1016/j.jss.2014.03.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 02/25/2014] [Accepted: 03/05/2014] [Indexed: 01/13/2023]
Abstract
BACKGROUND Systemic inflammation and increased matrix metalloproteinase (MMP) cause elastin degradation leading to abdominal aortic aneurysm (AAA) expansion. Several prospective studies report that statin therapy can reduce AAA expansion through anti-inflammation. We hypothesize that monocyte activity plays a pivotal role in this AAA development and this study examines patient peripheral blood monocyte cell adhesion, transendothelial migration, and MMP concentrations between AAA and non-AAA patients. MATERIALS AND METHODS Peripheral blood was collected and monocytes isolated from control (n=15) and AAA (n=13) patients. Monocyte adhesion, transmigration, and permeability assays were assessed. Luminex assays determined MMP-9 and tissue inhibitor of metalloproteinase-4 (TIMP-4) concentrations from cell culture supernatant and patient serum. RESULTS AAA patient monocytes showed increased adhesion to the endothelium relative fluorescence units (RFU, 0.33±0.17) versus controls (RFU, 0.13±0.04; P=0.005). Monocyte transmigration was also increased in AAA patients (RFU, 0.33±0.11) compared with controls (RFU, 0.25±0.04, P=0.01). Greater numbers of adhesive (R2=0.66) and transmigratory (R2=0.86) monocytes were directly proportional to the AAA diameter. Significantly higher serum levels of MMP-9 (2149.14±947 pg/mL) were found in AAA patients compared with controls (1189.2±293; P=0.01). TIMP-4 concentrations were significantly lower in AAA patients (826.7±100 pg/mL) compared with controls (1233±222 pg/mL; P=0.02). Cell culture supernatant concentrations of MMP and TIMP from cocultures were higher than monocyte-only cultures. CONCLUSIONS Monocytes from AAA patients have greater adhesion and transmigration through the endothelium in vitro, leading to elevated MMP-9 levels and the appropriate decrease in TIMP-4 levels. The ability to modulate monocyte activity may lead to novel medical therapies to decrease AAA expansion.
Collapse
Affiliation(s)
- Kiana M Samadzadeh
- Department of Research, Sacramento VA Medical Center, Mather, California
| | - Kevin C Chun
- Department of Research, Sacramento VA Medical Center, Mather, California
| | - Anthony T Nguyen
- Department of Research, Sacramento VA Medical Center, Mather, California
| | - Pamela M Baker
- Department of Research, Sacramento VA Medical Center, Mather, California
| | - Sukhmine Bains
- Department of Surgery, Sacramento VA Medical Center, Mather, California; Department of Surgery, University of California, Sacramento, California
| | - Eugene S Lee
- Department of Surgery, Sacramento VA Medical Center, Mather, California; Department of Surgery, University of California, Sacramento, California.
| |
Collapse
|
34
|
Zinnel NF, Russell DH. Size-to-charge dispersion of collision-induced dissociation product ions for enhancement of structural information and product ion identification. Anal Chem 2014; 86:4791-8. [PMID: 24754452 DOI: 10.1021/ac403929u] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ion mobility is used to disperse product ions formed by collision-induced dissociation (CID) on the basis of charge state and size-to-charge ratio. We previously described an approach for combining CID with ion mobility mass spectrometry (IM-MS) for dispersing fragment ions along charge state specific trend lines (Zinnel, N. F.; Pai, P. J.; Russell, D. H. Anal. Chem. 2012, 84, 3390; Sowell, R. A.; Koeniger, S. L.; Valentine, S. J.; Moon, M. H.; Clemmer, D. E. J. Am. Soc. Mass Spectrom. 2004, 15, 1341; McLean, J. A.; Ruotolo, B. T.; Gillig, K. J.; Russell, D. H. Int. J. Mass Spectrom. 2005, 240, 301), and this approach was used to assign metal ion binding sites for human metallothionein protein MT-2a (Chen, S. H.; Russell, W. K.; Russell, D. H. Anal. Chem. 2013, 85, 3229). Here, we use this approach to distinguish b-type N-terminal fragment ions from both internal fragment ions and y-type C-terminal fragment ions. We also show that in some cases specific secondary structural elements, viz., extended coils or helices, can be obtained for the y-type fragment ions series. The advantage of this approach is that product ion identity can be correlated to gas-phase ion structure, which provides rapid identification of the onset and termination of extended coil structure in peptides.
Collapse
Affiliation(s)
- Nathanael F Zinnel
- Laboratory for Biological Mass Spectrometry, Department of Chemistry, Texas A&M University , College Station, Texas 77842, United States
| | | |
Collapse
|
35
|
Madamanchi A, Santoro SA, Zutter MM. α2β1 Integrin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 819:41-60. [PMID: 25023166 DOI: 10.1007/978-94-017-9153-3_3] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The α2β1 integrin, also known as VLA-2, GPIa-IIa, CD49b, was first identified as an extracellular matrix receptor for collagens and/or laminins [55, 56]. It is now recognized that the α2β1 integrin serves as a receptor for many matrix and nonmatrix molecules [35, 79, 128]. Extensive analyses have clearly elucidated the α2 I domain structural motifs required for ligand binding, and also defined distinct conformations that lead to inactive, partially active or highly active ligand binding [3, 37, 66, 123, 136, 137, 140]. The mechanisms by which the α2β1 integrin plays a critical role in platelet function and homeostasis have been carefully defined via in vitro and in vivo experiments [76, 104, 117, 125]. Genetic and epidemiologic studies have confirmed human physiology and disease states mediated by this receptor in immunity, cancer, and development [6, 20, 21, 32, 43, 90]. The role of the α2β1 integrin in these multiple complex biologic processes will be discussed in the chapter.
Collapse
Affiliation(s)
- Aasakiran Madamanchi
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | |
Collapse
|
36
|
Cho J. Protein disulfide isomerase in thrombosis and vascular inflammation. J Thromb Haemost 2013; 11:2084-91. [PMID: 24118938 PMCID: PMC4076787 DOI: 10.1111/jth.12413] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Indexed: 12/14/2022]
Abstract
Protein disulfide isomerase (PDI) catalyzes disulfide bond oxidation, reduction and isomerization during protein synthesis in the endoplasmic reticulum (ER). In addition to its critical role in the ER, in vitro and in vivo studies with blocking antibodies and conditional knockout mice have demonstrated that cell surface PDI is required for thrombosis, hemostasis and vascular inflammation in a manner dependent on its isomerase activity. This review will focus on our current understanding of the pathophysiologic role of PDI in regulating integrin-mediated platelet and neutrophil functions during vascular disease.
Collapse
Affiliation(s)
- J Cho
- Departments of Pharmacology and Anesthesiology, University of Illinois College of Medicine, Chicago, IL, USA
| |
Collapse
|
37
|
Pepper LR, Parthasarathy R, Robbins GP, Dang NN, Hammer DA, Boder ET. Isolation of αL I domain mutants mediating firm cell adhesion using a novel flow-based sorting method. Protein Eng Des Sel 2013; 26:515-21. [PMID: 23788664 PMCID: PMC3711393 DOI: 10.1093/protein/gzt028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/07/2013] [Accepted: 05/09/2013] [Indexed: 11/12/2022] Open
Abstract
The inserted (I) domain of αLβ2 integrin (LFA-1) contains the entire binding site of the molecule. It mediates both rolling and firm adhesion of leukocytes at sites of inflammation depending on the activation state of the integrin. The affinity change of the entire integrin can be mimicked by the I domain alone through mutations that affect the conformation of the molecule. High-affinity mutants of the I domain have been discovered previously using both rational design and directed evolution. We have found that binding affinity fails to dictate the behavior of I domain adhesion under shear flow. In order to better understand I domain adhesion, we have developed a novel panning method to separate yeast expressing a library of I domain variants on the surface by adhesion under flow. Using conditions analogous to those experienced by cells interacting with the post-capillary vascular endothelium, we have identified mutations supporting firm adhesion that are not found using typical directed evolution techniques that select for tight binding to soluble ligands. Mutants isolated using this method do not cluster with those found by sorting with soluble ligand. Furthermore, these mutants mediate shear-driven cell rolling dynamics decorrelated from binding affinity, as previously observed for I domains bearing engineered disulfide bridges to stabilize activated conformational states. Characterization of these mutants supports a greater understanding of the structure-function relationship of the αL I domain, and of the relationship between applied force and bioadhesion in a broader context.
Collapse
Affiliation(s)
- Lauren R. Pepper
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Gregory P. Robbins
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas N. Dang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel A. Hammer
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eric T. Boder
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
38
|
Krishnan V, Dwivedi P, Kim BJ, Samal A, Macon K, Ma X, Mishra A, Doran KS, Ton-That H, Narayana SVL. Structure of Streptococcus agalactiae tip pilin GBS104: a model for GBS pili assembly and host interactions. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2013; 69:1073-89. [PMID: 23695252 PMCID: PMC3663123 DOI: 10.1107/s0907444913004642] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 02/18/2013] [Indexed: 11/10/2022]
Abstract
The crystal structure of a 75 kDa central fragment of GBS104, a tip pilin from the 2063V/R strain of Streptococcus agalactiae (group B streptococcus; GBS), is reported. In addition, a homology model of the remaining two domains of GBS104 was built and a model of full-length GBS104 was generated by combining the homology model (the N1 and N4 domains) and the crystal structure of the 75 kDa fragment (the N2 and N3 domains). This rod-shaped GBS104 model is constructed of three IgG-like domains (the N1, N2 and N4 domains) and one vWFA-like domain (the N3 domain). The N1 and N2 domains of GBS104 are assembled with distinct and remote segments contributed by the N- and C-termini. The metal-binding site in the N3 domain of GBS104 is in the closed/low-affinity conformation. Interestingly, this domain hosts two long arms that project away from the metal-binding site. Using site-directed mutagenesis, two cysteine residues that lock the N3 domain of GBS104 into the open/high-affinity conformation were introduced. Both wild-type and disulfide-locked recombinant proteins were tested for binding to extracellular matrix proteins such as collagen, fibronectin, fibrinogen and laminin, and an increase in fibronectin binding affinity was identified for the disulfide-locked N3 domain, suggesting that induced conformational changes may play a possible role in receptor binding.
Collapse
Affiliation(s)
- Vengadesan Krishnan
- UNESCO Regional Centre for Biotechnology (RCB), Gurgaon 122 016, Haryana, India
| | - Prabhat Dwivedi
- University of Texas Health Science Center, Houston, TX 77030, USA
| | - Brandon J. Kim
- Department of Biology and Center for Microbial Sciences, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Alexandra Samal
- Center for Biophysical Sciences and Engineering, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kevin Macon
- Center for Biophysical Sciences and Engineering, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Xin Ma
- University of Texas Health Science Center, Houston, TX 77030, USA
| | - Arunima Mishra
- University of Texas Health Science Center, Houston, TX 77030, USA
| | - Kelly S. Doran
- Department of Biology and Center for Microbial Sciences, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Hung Ton-That
- University of Texas Health Science Center, Houston, TX 77030, USA
| | - Sthanam V. L. Narayana
- Center for Biophysical Sciences and Engineering, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
39
|
Pérez-Rivero G, Cascio G, Soriano SF, Sanz ÁG, de Guinoa JS, Rodríguez-Frade JM, Gomariz RP, Holgado BL, Cabañas C, Carrasco YR, Stein JV, Mellado M. Janus kinases 1 and 2 regulate chemokine-mediated integrin activation and naïve T-cell homing. Eur J Immunol 2013; 43:1745-57. [PMID: 23526587 DOI: 10.1002/eji.201243178] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 02/08/2013] [Accepted: 03/19/2013] [Indexed: 12/21/2022]
Abstract
Janus kinases (JAKs) are central signaling molecules in cytokine receptor cascades. Although they have also been implicated in chemokine receptor signaling, this function continues to be debated. To address this issue, we established a nucleofection model in primary, nonactivated mouse T lymphocytes to silence JAK expression and to evaluate the ability of these cells to home to lymph nodes. Reduced JAK1 and JAK2 expression impaired naïve T-cell migration in response to gradients of the chemokines CXCL12 and CCL21. In vivo homing of JAK1/JAK2-deficient cells to lymph nodes decreased, whereas intranodal localization and motility were unaffected. JAK1 and JAK2 defects altered CXCL12- and CCL21-triggered ezrin/radixin/moesin (ERM) dephosphorylation and F-actin polymerization, as well as activation of lymphocyte function-associated Ag-1 and very late Ag-4 integrins. As a result, the cells did not adhere firmly to integrin substrates in response to these chemokines. The results demonstrate that JAK1/JAK2 participate in chemokine-induced integrin activation and might be considered a target for modulation of immune cell extravasation and therefore, control of inflammatory reactions.
Collapse
Affiliation(s)
- Gema Pérez-Rivero
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Caspases are a powerful class of cysteine proteases. Introduction of activated caspases in healthy or cancerous cells results in induction of apoptotic cell death. In this study, we have designed and characterized a version of caspase-7 that can be inactivated under oxidizing extracellular conditions and then reactivated under reducing intracellular conditions. This version of caspase-7 is allosterically inactivated when two of the substrate-binding loops are locked together via an engineered disulfide. When this disulfide is reduced, the protein regains its full function. The inactive loop-locked version of caspase-7 can be readily observed by immunoblotting and mass spectrometry. The reduced and reactivated form of the enzyme observed crystallographically is the first caspase-7 structure in which the substrate-binding groove is properly ordered even in the absence of an active-site ligand. In the reactivated structure, the catalytic-dyad cysteine-histidine are positioned 3.5 Å apart in an orientation that is capable of supporting catalysis. This redox-controlled version of caspase-7 is particularly well suited for targeted cell death in concert with redox-triggered delivery vehicles.
Collapse
Affiliation(s)
- Witold A Witkowski
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, USA
| | | |
Collapse
|
41
|
Extracellular protein disulfide isomerase regulates ligand-binding activity of αMβ2 integrin and neutrophil recruitment during vascular inflammation. Blood 2013; 121:3789-800, S1-15. [PMID: 23460613 DOI: 10.1182/blood-2012-11-467985] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
β2 integrins play a crucial role during neutrophil recruitment into the site of vascular inflammation. However, it remains unknown how ligand-binding activity of the integrin is regulated. Using fluorescence intravital microscopy in mice generated by crossing protein disulfide isomerase (PDI) floxed mice with lysozyme-Cre transgenic mice, we demonstrate that neutrophil PDI is required for neutrophil adhesion and crawling during tumor necrosis factor-α-induced vascular inflammation in vivo. Rescue experiments show that the isomerase activity of extracellular PDI is critical for its regulatory effect on neutrophil recruitment. Studies with blocking anti-PDI antibodies and αLβ2 or αMβ2 null mice suggest that extracellular PDI regulates αMβ2 integrin-mediated adhesive function of neutrophils during vascular inflammation. Consistently, we show that neutrophil surface PDI is important for αMβ2 integrin-mediated adhesion of human neutrophils under shear and static conditions and for binding of soluble fibrinogen to activated αMβ2 integrin. Confocal microscopy and biochemical studies reveal that neutrophil surface PDI interacts with αMβ2 integrin in lipid rafts of stimulated neutrophils and regulates αMβ2 integrin clustering, presumably by changing the redox state of the integrin. Thus, our results provide the first evidence that extracellular PDI could be a novel therapeutic target for preventing and treating inappropriate neutrophil sequestration.
Collapse
|
42
|
San Sebastián E, Zimmerman T, Zubia A, Vara Y, Martin E, Sirockin F, Dejaegere A, Stote RH, Lopez X, Pantoja-Uceda D, Valcárcel M, Mendoza L, Vidal-Vanaclocha F, Cossío FP, Blanco FJ. Design, synthesis, and functional evaluation of leukocyte function associated antigen-1 antagonists in early and late stages of cancer development. J Med Chem 2013; 56:735-47. [PMID: 23339734 DOI: 10.1021/jm3016848] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The integrin leukocyte function associated antigen 1 (LFA-1) binds the intercellular adhesion molecule 1 (ICAM-1) by its α(L)-chain inserted domain (I-domain). This interaction plays a key role in cancer and other diseases. We report the structure-based design, small-scale synthesis, and biological activity evaluation of a novel family of LFA-1 antagonists. The design led to the synthesis of a family of highly substituted homochiral pyrrolidines with antiproliferative and antimetastatic activity in a murine model of colon carcinoma, as well as potent antiadhesive properties in several cancer cell lines in the low micromolar range. NMR analysis of their binding to the isolated I-domain shows that they bind to the I-domain allosteric site (IDAS), the binding site of other allosteric LFA-1 inhibitors. These results provide evidence of the potential therapeutic value of a new set of LFA-1 inhibitors, whose further development is facilitated by a synthetic strategy that is versatile and fully stereocontrolled.
Collapse
|
43
|
Zhu C, Jiang N, Huang J, Zarnitsyna VI, Evavold BD. Insights from in situ analysis of TCR-pMHC recognition: response of an interaction network. Immunol Rev 2013; 251:49-64. [PMID: 23278740 PMCID: PMC3539230 DOI: 10.1111/imr.12016] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recognition of peptide presented by the major histocompatibility complex (pMHC) molecule by the T-cell receptor (TCR) determines T-cell selection, development, differentiation, fate, and function. Despite intensive studies on the structures, thermodynamic properties, kinetic rates, and affinities of TCR-pMHC interactions in the past two decades, questions regarding the functional outcome of these interactions, i.e. how binding of the αβ TCR heterodimer with distinct pMHCs triggers different intracellular signals via the adjacent CD3 components to produce different T-cell responses, remain unclear. Most kinetic measurements have used surface plasmon resonance, a three-dimensional (3D) technique in which fluid-phase receptors and ligands are removed from their cellular environment. Recently, several two-dimensional (2D) techniques have been developed to analyze molecular interactions on live T cells with pMHCs presented by surrogate antigen-presenting cells or supported planar lipid bilayers. The insights from these in situ analyses have provided a sharp contrast of the 2D network biology approach to the 3D reductionist approach and prompted rethinking of our current views of T-cell triggering. Based on these insights, we propose a mechanochemical coupled triggering hypothesis to explain why the in situ kinetic parameters differ so much from their 3D counterparts, yet correlate so much better with T-cell functional responses.
Collapse
Affiliation(s)
- Cheng Zhu
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA.
| | | | | | | | | |
Collapse
|
44
|
Tanigami H, Okamoto T, Yasue Y, Shimaoka M. Astroglial integrins in the development and regulation of neurovascular units. PAIN RESEARCH AND TREATMENT 2012; 2012:964652. [PMID: 23304493 PMCID: PMC3529429 DOI: 10.1155/2012/964652] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 11/13/2012] [Indexed: 11/18/2022]
Abstract
In the neurovascular units of the central nervous system, astrocytes form extensive networks that physically and functionally connect the neuronal synapses and the cerebral vascular vessels. This astrocytic network is thought to be critically important for coupling neuronal signaling activity and energy demand with cerebral vascular tone and blood flow. To establish and maintain this elaborate network, astrocytes must precisely calibrate their perisynaptic and perivascular processes in order to sense and regulate neuronal and vascular activities, respectively. Integrins, a prominent family of cell-adhesion molecules that support astrocytic migration in the brain during developmental and normal adult stages, have been implicated in regulating the integrity of the blood brain barrier and the tripartite synapse to facilitate the formation of a functionally integrated neurovascular unit. This paper describes the significant roles that integrins and connexins play not only in regulating astrocyte migration during the developmental and adult stages of the neurovascular unit, but also in general health and in such diseases as hepatic encephalopathy.
Collapse
Affiliation(s)
- Hironobu Tanigami
- Department of Anesthesiology, Osaka Medical Center for Cancer and Cardiovascular Diseases, 1-3-3 Nakamichi, Higashinari-ku, Osaka 537-8511, Japan
| | - Takayuki Okamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Mie, Tsu City, Japan
| | - Yuichi Yasue
- Department of Anesthesiology, Osaka Medical Center for Cancer and Cardiovascular Diseases, 1-3-3 Nakamichi, Higashinari-ku, Osaka 537-8511, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Mie, Tsu City, Japan
| |
Collapse
|
45
|
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
|
46
|
Abstract
Integrins are a family of α/β heterodimeric adhesion metalloprotein receptors and their functions are highly dependent on and regulated by different divalent cations. Recently advanced studies have revolutionized our perception of integrin metal ion-binding sites and their specific functions. Ligand binding to integrins is bridged by a divalent cation bound at the MIDAS motif on top of either α I domain in I domain-containing integrins or β I domain in α I domain-less integrins. The MIDAS motif in β I domain is flanked by ADMIDAS and SyMBS, the other two crucial metal ion binding sites playing pivotal roles in the regulation of integrin affinity and bidirectional signaling across the plasma membrane. The β-propeller domain of α subunit contains three or four β-hairpin loop-like Ca(2+)-binding motifs that have essential roles in integrin biogenesis. The function of another Ca(2+)-binding motif located at the genu of α subunit remains elusive. Here, we provide an overview of the integrin metal ion-binding sites and discuss their roles in the regulation of integrin functions.
Collapse
Affiliation(s)
- Kun Zhang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | |
Collapse
|
47
|
Zarnitsyna V, Zhu C. T cell triggering: insights from 2D kinetics analysis of molecular interactions. Phys Biol 2012; 9:045005. [PMID: 22871794 DOI: 10.1088/1478-3975/9/4/045005] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Interaction of the T cell receptor (TCR) with pathogen-derived peptide presented by the major histocompatibility complex (pMHC) molecule is central to adaptive immunity as it initiates intracellular signaling to trigger T cell response to infection. Kinetic parameters of this interaction have been under intensive investigation for more than two decades using soluble pMHCs and/or TCRs with at least one of them in the solution (three-dimensional (3D) methods). Recently, several techniques have been developed to enable kinetic analysis on live T cells with pMHCs presented by surrogate antigen presenting cells (APCs) or supported planar lipid bilayers (two-dimensional (2D) methods). Comparison of 2D versus 3D parameters reveals drastic differences with broader ranges of 2D affinities and on-rates and orders of magnitude faster 2D off-rates for functionally distinct pMHCs. Here we review new 2D data and discuss how it may impact previously developed models of T cell discrimination between pMHCs of different potencies.
Collapse
Affiliation(s)
- Veronika Zarnitsyna
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA.
| | | |
Collapse
|
48
|
Chen G, Dimitriou I, Milne L, Lang KS, Lang PA, Fine N, Ohashi PS, Kubes P, Rottapel R. The 3BP2 adapter protein is required for chemoattractant-mediated neutrophil activation. THE JOURNAL OF IMMUNOLOGY 2012; 189:2138-50. [PMID: 22815290 DOI: 10.4049/jimmunol.1103184] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
3BP2 is a pleckstrin homology and Src homology 2 domain-containing adapter protein mutated in cherubism, a rare autosomal-dominant human bone disorder. Previously, we have demonstrated a functional role for 3BP2 in peripheral B cell development and in peritoneal B1 and splenic marginal zone B cell-mediated Ab responses. In this study, we show that 3BP2 is required for G protein-coupled receptor-mediated neutrophil functions. Neutrophils derived from 3BP2-deficient (Sh3bp2-/-) mice failed to polarize their actin cytoskeleton or migrate in response to a gradient of chemotactic peptide, fMLF. Sh3bp2-/- neutrophils failed to adhere, crawl, and emigrate out of the vasculature in response to fMLF superfusion. 3BP2 is required for optimal activation of Src family kinases, small GTPase Rac2, neutrophil superoxide anion production, and for Listeria monocytogenes bacterial clearance in vivo. The functional defects observed in Sh3bp2-/- neutrophils may partially be explained by the failure to fully activate Vav1 guanine nucleotide exchange factor and properly localize P-Rex1 guanine nucleotide exchange factor at the leading edge of migrating cells. Our results reveal an obligate requirement for the adapter protein 3BP2 in G protein-coupled receptor-mediated neutrophil function.
Collapse
Affiliation(s)
- Grace Chen
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 148, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
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
|
50
|
Abstract
Cell adhesion depends on combinational expression and interactions of a large number of adhesion molecules at cell-to-cell or cell-to-matrix contact sites. Integrins and their immunoglobulin superfamily (IgSF) ligands represent foremost classes of cell adhesion molecules in immune system. Structural study is critical for a better understanding of the interactions between integrins and their IgSF ligands. Here we describe protocols for protein expression of integrin αL I domain and its IgSF ligand ICAM-5 D1D2 fragment for crystallography.
Collapse
|