1
|
Yang G, Alarcon C, Chanfreau C, Lee NH, Friedman P, Nutescu E, Tuck M, O'Brien T, Gong L, Klein TE, Chang K, Tsao PS, Meltzer DO, Lynch JA, Tuteja S, Perera MA. Investigation of Genomic and Transcriptomic Risk Factors of Clopidogrel Response in African Americans. Clin Pharmacol Ther 2025; 117:1313-1324. [PMID: 39868839 PMCID: PMC11993291 DOI: 10.1002/cpt.3552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 12/17/2024] [Indexed: 01/28/2025]
Abstract
Clopidogrel, an anti-platelet drug, is used to prevent thrombosis after percutaneous coronary intervention. Clopidogrel resistance results in recurring ischemic events, with African Americans (AA) suffering disproportionately. The aim of this study was to discover novel biomarkers of clopidogrel resistance in African Americans using genome and transcriptome data. We conducted a genome-wide association study (GWAS), including local ancestry adjustment, in 141 AA on clopidogrel to identify genetic associations with high on-treatment platelet reactivity (HTPR), with validation of genome-wide significant and suggestive loci in an independent cohort of AA clopidogrel patients (N = 823) from the Million Veteran's Program (MVP) along with in vitro functional analysis. We performed differential gene expression (DGE) analysis in whole blood to identify transcriptomic predictors of response, followed by functional validation in MEG-01 cells. GWAS identified one signal on Chromosome 7 as significantly associated with increasing risk of HTPR. The lead single-nucleotide polymorphism (SNP), rs7807369, within thrombospondin 7A (THSD7A) was associated with an increased risk of HTPR (odds ratio (OR) = 4.02, P = 4.56 × 10-9). Higher THSD7A gene expression was associated with HTPR in an independent cohort of clopidogrel-treated patients (P = 0.004) and carrying a risk allele showed increased gene expression in primary human endothelial cells. Notably, the CYP2C19*2 variants showed no association with clopidogrel response in the discovery or MVP cohorts. DGE analysis identified an association with decreased LAIR1 and AP3B2 expression to HTPR. LAIR1 knockdown in MEG-01 cells resulted in increased expression of SYK and AKT1, suggesting an inhibitory role of LAIR1 in the Glycoprotein VI pathway. In summary, these findings suggest that other variants and genes outside of CYP2C19 star alleles play an important role in clopidogrel response in AA.
Collapse
Affiliation(s)
- Guang Yang
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Center for Applied BioinformaticsSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - Cristina Alarcon
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Catherine Chanfreau
- VA Informatics and Computing Infrastructure (VINCI)VA Salt Lake City Health Care SystemSalt Lake CityUtahUSA
| | - Norman H. Lee
- Department of Pharmacology and PhysiologyGeorge Washington UniversityWashingtonDCUSA
- GW Cancer CenterGeorge Washington UniversityWashington, DCUSA
| | - Paula Friedman
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Edith Nutescu
- Department of Pharmacy PracticeRetzky College of Pharmacy, University of Illinois ChicagoChicagoIllinoisUSA
| | - Matthew Tuck
- Washington DC VA Medical CenterWashingtonDCUSA
- The George Washington UniversityWashingtonDCUSA
| | - Travis O'Brien
- Department of Pharmacology and PhysiologyGeorge Washington UniversityWashingtonDCUSA
| | - Li Gong
- Department of Biomedical Data ScienceStanford UniversityStanfordCaliforniaUSA
| | - Teri E. Klein
- Department of Biomedical Data Science and Department of MedicineStanford UniversityStanfordCaliforniaUSA
| | - Kyong‐Mi Chang
- Corporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Philip S. Tsao
- VA Palo Alto Healthcare System and Stanford UniversityPalo AltoCaliforniaUSA
| | - David O. Meltzer
- Section of Hospital Medicine, Department of MedicineUniversity of ChicagoChicagoIllinoisUSA
| | - Julie A. Lynch
- VA Informatics and Computing Infrastructure (VINCI)VA Salt Lake City Health Care SystemSalt Lake CityUtahUSA
| | | | - Sony Tuteja
- Corporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Minoli A. Perera
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
2
|
Malcor JD, Ferruz N, Romero-Romero S, Dhingra S, Sagar V, Jalan AA. Deciphering the folding code of collagens. Nat Commun 2025; 16:2702. [PMID: 40108160 PMCID: PMC11923368 DOI: 10.1038/s41467-024-54046-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 10/30/2024] [Indexed: 03/22/2025] Open
Abstract
Collagen proteins contain a characteristic structural motif called a triple helix. During the self-assembly of this motif, three polypeptides form a folding nucleus at the C-termini and then propagate towards the N-termini like a zip-chain. While polypeptides from human collagens contain up to a 1000 amino acids, those found in bacteria can contain up to 6000 amino acids. Additionally, the collagen polypeptides are also frequently interrupted by non-helical sequences that disrupt folding and reduce stability. Given the length of polypeptides and the disruptive interruptions, compensating mechanisms that stabilize against local unfolding during propagation and offset the entropic cost of folding are not fully understood. Here, we show that the information for the correct folding of collagen triple helices is encoded in their sequence as interchain electrostatic interactions, which likely act as molecular clamps that prevent local unfolding. In the case of humans, disrupting these electrostatic interactions is associated with severe to lethal diseases.
Collapse
Affiliation(s)
- Jean-Daniel Malcor
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305 University of Lyon, Lyon, France
| | - Noelia Ferruz
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
- Centre for Genomic Regulation, Barcelona, Spain
| | - Sergio Romero-Romero
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
- Department of Biochemistry and Structural Biology. Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Surbhi Dhingra
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
| | - Vamika Sagar
- Department of Biomaterials, University of Bayreuth, Bayreuth, Germany
| | - Abhishek A Jalan
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany.
- Department of Biomaterials, University of Bayreuth, Bayreuth, Germany.
| |
Collapse
|
3
|
Carvalheiro T, Marut W, Pascoal Ramos MI, García S, Fleury D, Affandi AJ, Meijers AS, Giovannone B, Tieland RG, Elshof E, Ottria A, Cossu M, Meizlish ML, Veenendaal T, Ramanujam M, Moreno-García ME, Klumperman J, Liv N, Radstake TRDJ, Meyaard L. Impaired LAIR-1-mediated immune control due to collagen degradation in fibrosis. J Autoimmun 2024; 146:103219. [PMID: 38696927 DOI: 10.1016/j.jaut.2024.103219] [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: 11/06/2023] [Revised: 03/10/2024] [Accepted: 03/29/2024] [Indexed: 05/04/2024]
Abstract
Tissue repair is disturbed in fibrotic diseases like systemic sclerosis (SSc), where the deposition of large amounts of extracellular matrix components such as collagen interferes with organ function. LAIR-1 is an inhibitory collagen receptor highly expressed on tissue immune cells. We questioned whether in SSc, impaired LAIR-1-collagen interaction is contributing to the ongoing inflammation and fibrosis. We found that SSc patients do not have an intrinsic defect in LAIR-1 expression or function. Instead, fibroblasts from healthy controls and SSc patients stimulated by soluble factors that drive inflammation and fibrosis in SSc deposit disorganized collagen products in vitro, which are dysfunctional LAIR-1 ligands. This is dependent of matrix metalloproteinases and platelet-derived growth factor receptor signaling. In support of a non-redundant role of LAIR-1 in the control of fibrosis, we found that LAIR-1-deficient mice have increased skin fibrosis in response to repeated injury and in the bleomycin mouse model for SSc. Thus, LAIR-1 represents an essential control mechanism for tissue repair. In fibrotic disease, excessive collagen degradation may lead to a disturbed feedback loop. The presence of functional LAIR-1 in patients provides a therapeutic opportunity to reactivate this intrinsic negative feedback mechanism in fibrotic diseases.
Collapse
Affiliation(s)
- Tiago Carvalheiro
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Wioleta Marut
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - M Inês Pascoal Ramos
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Samuel García
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Devan Fleury
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim, Ridgefield, USA
| | - Alsya J Affandi
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Aniek S Meijers
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Barbara Giovannone
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Dermatology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ralph G Tieland
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Eline Elshof
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Andrea Ottria
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Marta Cossu
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Matthew L Meizlish
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, USA
| | - Tineke Veenendaal
- Cell Biology, Centre for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Meera Ramanujam
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim, Ridgefield, USA
| | | | - Judith Klumperman
- Cell Biology, Centre for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Nalan Liv
- Cell Biology, Centre for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Timothy R D J Radstake
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands.
| |
Collapse
|
4
|
Yang G, Alarcon C, Chanfreau C, Lee NH, Friedman P, Nutescu E, Tuck M, O'Brien T, Gong L, Klein TE, Chang KM, Tsao PS, Meltzer DO, Tuteja S, Perera MA. Investigation of genomic and transcriptomic risk factors in clopidogrel response in African Americans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.05.23299140. [PMID: 38106031 PMCID: PMC10723512 DOI: 10.1101/2023.12.05.23299140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Clopidogrel, an anti-platelet drug, used to prevent thrombosis after percutaneous coronary intervention. Clopidogrel resistance results in recurring ischemic episodes, with African Americans suffering disproportionately. The aim of this study was to identify biomarkers of clopidogrel resistance in African American patients. We conducted a genome-wide association study, including local ancestry adjustment, in 141 African Americans on clopidogrel to identify associations with high on-treatment platelet reactivity (HTPR). We validated genome-wide and suggestive hits in an independent cohort of African American clopidogrel patients (N = 823) from the Million Veteran's Program (MVP) along with in vitro functional follow up. We performed differential gene expression (DGE) analysis in whole blood with functional follow-up in MEG-01 cells. We identified rs7807369, within thrombospondin 7A (THSD7A), as significantly associated with increasing risk of HTPR (p = 4.56 × 10-9). Higher THSD7A expression was associated with HTPR in an independent gene expression cohort of clopidogrel treated patients (p = 0.004) and supported by increased gene expression on THSD7A in primary human endothelial cells carrying the risk haplotype. Two SNPs (rs1149515 and rs191786) were validated in the MVP cohort. DGE analysis identified an association with decreased LAIR1 expression to HTPR. LAIR1 knockdown in a MEG-01 cells resulted in increased expression of SYK and AKT1, suggesting an inhibitory role of LAIR1 in the Glycoprotein VI pathway. Notably, the CYP2C19 variants showed no association with clopidogrel response in the discovery or MVP cohorts. In summary, these finding suggest that other variants outside of CYP2C19 star alleles play an important role in clopidogrel response in African Americans.
Collapse
Affiliation(s)
- Guang Yang
- Department of Pharmacology, Center for Pharmacogenomics, Fienberg School of Medicine, Northwestern University, Chicago IL
| | - Cristina Alarcon
- Department of Pharmacology, Center for Pharmacogenomics, Fienberg School of Medicine, Northwestern University, Chicago IL
| | | | - Norman H Lee
- Department of Pharmacology and Physiology, George Washington University, 2300 I Street NW, Washington, DC, 20037, USA
| | - Paula Friedman
- Department of Pharmacology, Center for Pharmacogenomics, Fienberg School of Medicine, Northwestern University, Chicago IL
| | - Edith Nutescu
- Department of Pharmacy Practice and Center for Pharmacoepidemiology and Pharmacoeconomic Research, University of Illinois Chicago, College of Pharmacy, Chicago, IL
| | - Matthew Tuck
- Washington DC VA Medical Center, Washington, DC and The George Washington University, Washington, DC
| | - Travis O'Brien
- Department of Pharmacology and Physiology, George Washington University, 2300 I Street NW, Washington, DC, 20037, USA
| | - Li Gong
- Department of Biomedical Data Science, Stanford University, Stanford, CA
| | - Teri E Klein
- Department of Biomedical Data Science and Department of Medicine, Stanford University, Stanford, CA
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Philip S Tsao
- VA Palo Alto Healthcare System and Stanford University, Palo Alto, CA
| | - David O Meltzer
- Section of Hospital Medicine, Department of Medicine, University of Chicago, Chicago, IL
| | - Sony Tuteja
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Minoli A Perera
- Department of Pharmacology, Center for Pharmacogenomics, Fienberg School of Medicine, Northwestern University, Chicago IL
| |
Collapse
|
5
|
Ye Y, Yang L, Leng M, Wang Q, Wu J, Wan W, Wang H, Li L, Peng Y, Chai S, Meng Z. Luteolin inhibits GPVI-mediated platelet activation, oxidative stress, and thrombosis. Front Pharmacol 2023; 14:1255069. [PMID: 38026984 PMCID: PMC10644720 DOI: 10.3389/fphar.2023.1255069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction: Luteolin inhibits platelet activation and thrombus formation, but the mechanisms are unclear. This study investigated the effects of luteolin on GPVI-mediated platelet activation in vitro and explored the effect of luteolin on thrombosis, coagulation, and platelet production in vivo. Methods: Washed human platelets were used for aggregation, membrane protein expression, ATP, Ca2+, and LDH release, platelet adhesion/spreading, and clot retraction experiments. Washed human platelets were used to detect collagen and convulxin-induced reactive oxygen species production and endogenous antioxidant effects. C57BL/6 male mice were used for ferric chloride-induced mesenteric thrombosis, collagen-epinephrine induced acute pulmonary embolism, tail bleeding, coagulation function, and luteolin toxicity experiments. The interaction between luteolin and GPVI was analyzed using solid phase binding assay and surface plasmon resonance (SPR). Results: Luteolin inhibited collagen- and convulxin-mediated platelet aggregation, adhesion, and release. Luteolin inhibited collagen- and convulxin-induced platelet ROS production and increased platelet endogenous antioxidant capacity. Luteolin reduced convulxin-induced activation of ITAM and MAPK signaling molecules. Molecular docking simulation showed that luteolin forms hydrogen bonds with GPVI. The solid phase binding assay showed that luteolin inhibited the interaction between collagen and GPVI. Surface plasmon resonance showed that luteolin bonded GPVI. Luteolin inhibited integrin αIIbβ3-mediated platelet activation. Luteolin inhibited mesenteric artery thrombosis and collagen- adrenergic-induced pulmonary thrombosis in mice. Luteolin decreased oxidative stress in vivo. Luteolin did not affect coagulation, hemostasis, or platelet production in mice. Discussion: Luteolin may be an effective and safe antiplatelet agent target for GPVI. A new mechanism (decreased oxidative stress) for the anti-platelet activity of luteolin has been identified.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Zhaohui Meng
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
6
|
Tripathi AS, Zaki MEA, Al-Hussain SA, Dubey BK, Singh P, Rind L, Yadav RK. Material matters: exploring the interplay between natural biomaterials and host immune system. Front Immunol 2023; 14:1269960. [PMID: 37936689 PMCID: PMC10627157 DOI: 10.3389/fimmu.2023.1269960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/02/2023] [Indexed: 11/09/2023] Open
Abstract
Biomaterials are widely used for various medical purposes, for instance, implants, tissue engineering, medical devices, and drug delivery systems. Natural biomaterials can be obtained from proteins, carbohydrates, and cell-specific sources. However, when these biomaterials are introduced into the body, they trigger an immune response which may lead to rejection and failure of the implanted device or tissue. The immune system recognizes natural biomaterials as foreign substances and triggers the activation of several immune cells, for instance, macrophages, dendritic cells, and T cells. These cells release pro-inflammatory cytokines and chemokines, which recruit other immune cells to the implantation site. The activation of the immune system can lead to an inflammatory response, which can be beneficial or detrimental, depending on the type of natural biomaterial and the extent of the immune response. These biomaterials can also influence the immune response by modulating the behavior of immune cells. For example, biomaterials with specific surface properties, such as charge and hydrophobicity, can affect the activation and differentiation of immune cells. Additionally, biomaterials can be engineered to release immunomodulatory factors, such as anti-inflammatory cytokines, to promote a tolerogenic immune response. In conclusion, the interaction between biomaterials and the body's immune system is an intricate procedure with potential consequences for the effectiveness of therapeutics and medical devices. A better understanding of this interplay can help to design biomaterials that promote favorable immune responses and minimize adverse reactions.
Collapse
Affiliation(s)
| | - Magdi E A Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad lbn Saud Islamic University, Riyadh, Saudi Arabia
| | - Sami A Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad lbn Saud Islamic University, Riyadh, Saudi Arabia
| | - Bidhyut Kumar Dubey
- Department of Pharmaceutical Chemistry, Era College of Pharmacy, Era University, Lucknow, India
| | - Prabhjot Singh
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, India
| | - Laiba Rind
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, India
| | - Rajnish Kumar Yadav
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, India
| |
Collapse
|
7
|
Wang J, Hu H, Wang J, Qiu H, Gao Y, Xu Y, Liu Z, Tang Y, Song L, Ramshaw J, Lin H, Zhang X. Characterization of recombinant humanized collagen type III and its influence on cell behavior and phenotype. JOURNAL OF LEATHER SCIENCE AND ENGINEERING 2022. [DOI: 10.1186/s42825-022-00103-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
AbstractCollagen made a tremendous impact in the field of regenerative medicine as a bioactive material. For decades, collagen has been used not only as a scaffolding material but also as an active component in regulating cells' biological behavior and phenotype. However, animal-derived collagen as a major source suffered from problems of immunogenicity, risk of viral infection, and the unclear relationship between bioactive sequence and function. Recombinant humanized collagen (rhCol) provided alternatives for regenerative medicine with more controllable risks. However, the characterization of rhCol and the interaction between rhCol and cells still need further investigation, including cell behavior and phenotype. The current study preliminarily demonstrated that recombinant humanized collagen type III (rhCol III) conformed to the theoretical amino acid sequence and had an advanced structure resembling bovine collagen. Furthermore, rhCol III could facilitate basal biological behaviors of human skin fibroblasts, such as adhesion, proliferation and migration. rhCol III was beneficial for some extracellular matrix-expressing cell phenotypes. The study would shed light on the mechanism research of rhCol and cell interactions and further understanding of effectiveness in tissue regeneration.
Graphical abstract
Collapse
|
8
|
Xie J, Gui X, Deng M, Chen H, Chen Y, Liu X, Ku Z, Tan L, Huang R, He Y, Zhang B, Lewis C, Chen K, Xu L, Xu J, Huang T, Liao XC, Zhang N, An Z, Zhang CC. Blocking LAIR1 signaling in immune cells inhibits tumor development. Front Immunol 2022; 13:996026. [PMID: 36211388 PMCID: PMC9534319 DOI: 10.3389/fimmu.2022.996026] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/01/2022] [Indexed: 12/31/2022] Open
Abstract
The current immune checkpoint blockade therapy has been successful in treating some cancers but not others. New molecular targets and therapeutic approaches of cancer immunology need to be identified. Leukocyte associated immunoglobulin like receptor 1 (LAIR1) is an immune inhibitory receptor expressing on most immune cell types. However, it remains a question whether we can specifically and actively block LAIR1 signaling to activate immune responses for cancer treatment. Here we report the development of specific antagonistic anti-LAIR1 monoclonal antibodies and studied the effects of LAIR1 blockade on the anti-tumor immune functions. The anti-LAIR1 antagonistic antibody stimulated the activities of T cells, natural killer cells, macrophages, and dendritic cells in vitro. The single-cell RNA sequencing analysis of intratumoral immune cells in syngeneic human LAIR1 transgenic mice treated with control or anti-LAIR1 antagonist antibodies indicates that LAIR1 signaling blockade increased the numbers of CD4 memory T cells and inflammatory macrophages, but decreased those of pro-tumor macrophages, regulatory T cells, and plasmacytoid dendritic cells. Importantly, the LAIR1 blockade by the antagonistic antibody inhibited the activity of immunosuppressive myeloid cells and reactivated T cells from cancer patients in vitro and impeded tumor metastasis in a humanized mouse model. Blocking LAIR1 signaling in immune cells represents a promising strategy for development of anti-cancer immunotherapy.
Collapse
Affiliation(s)
- Jingjing Xie
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Xun Gui
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX, United States
| | - Mi Deng
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Heyu Chen
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yuanzhi Chen
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX, United States
| | - Xiaoye Liu
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX, United States
| | - Lingxiao Tan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX, United States
| | - Ryan Huang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yubo He
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Bruce Zhang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Cheryl Lewis
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Kenian Chen
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pediatrics, University of Texas Southwestern Medical Center,
Dallas, TX, United States
| | - Lin Xu
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pediatrics, University of Texas Southwestern Medical Center,
Dallas, TX, United States
| | - Jian Xu
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Tao Huang
- Immune-Onc Therapeutics, Inc, Palo Alto, CA, United States
| | | | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX, United States
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX, United States
| | - Cheng Cheng Zhang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
9
|
Malcor JD, Mallein-Gerin F. Biomaterial functionalization with triple-helical peptides for tissue engineering. Acta Biomater 2022; 148:1-21. [PMID: 35675889 DOI: 10.1016/j.actbio.2022.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/09/2022] [Accepted: 06/01/2022] [Indexed: 11/29/2022]
Abstract
In the growing field of tissue engineering, providing cells in biomaterials with the adequate biological cues represents an increasingly important challenge. Yet, biomaterials with excellent mechanical properties often are often biologically inert to many cell types. To address this issue, researchers resort to functionalization, i.e. the surface modification of a biomaterial with active molecules or substances. Functionalization notably aims to replicate the native cellular microenvironment provided by the extracellular matrix, and in particular by collagen, its major component. As our understanding of biological processes regulating cell behaviour increases, functionalization with biomolecules binding cell surface receptors constitutes a promising strategy. Amongst these, triple-helical peptides (THPs) that reproduce the architectural and biological properties of collagen are especially attractive. Indeed, THPs containing binding sites from the native collagen sequence have successfully been used to guide cell response by establishing cell-biomaterial interactions. Notably, the GFOGER motif recognising the collagen-binding integrins is extensively employed as a cell adhesive peptide. In biomaterials, THPs efficiently improved cell adhesion, differentiation and function on biomaterials designed for tissue repair (especially for bone, cartilage, tendon and heart), vascular graft fabrication, wound dressing, drug delivery or immunomodulation. This review describes the key characteristics of THPs, their effect on cells when combined to biomaterials and their strong potential as biomimetic tools for regenerative medicine. STATEMENT OF SIGNIFICANCE: This review article describes how triple-helical peptides constitute efficient tools to improve cell-biomaterial interactions in tissue engineering. Triple helical peptides are bioactive molecules that mimic the architectural and biological properties of collagen. They have been successfully used to specifically recognize cell-surface receptors and provide cells seeded on biomaterials with controlled biological cues. Functionalization with triple-helical peptides has enabled researchers to improve cell function for regenerative medicine applications, such as tissue repair. However, despite encouraging results, this approach remains limited and under-exploited, and most functionalization strategies reported in the literature rely on biomolecules that are unable to address collagen-binding receptors. This review will assist researchers in selecting the correct tools to functionalize biomaterials in efforts to guide cellular response.
Collapse
Affiliation(s)
- Jean-Daniel Malcor
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, Cedex 07, Lyon 69367, France.
| | - Frédéric Mallein-Gerin
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, Cedex 07, Lyon 69367, France
| |
Collapse
|
10
|
GPVI and collagen: the final word? Blood 2022; 139:3005-3007. [PMID: 35587869 DOI: 10.1182/blood.2022015962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 02/26/2022] [Indexed: 11/20/2022] Open
|
11
|
Structural insights into collagen binding by platelet receptor glycoprotein VI. Blood 2022; 139:3087-3098. [PMID: 35245360 DOI: 10.1182/blood.2021013614] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 02/08/2022] [Indexed: 02/02/2023] Open
Abstract
Glycoprotein VI (GPVI) mediates collagen-induced platelet activation after vascular damage and is an important contributor to the onset of thrombosis, heart attack, and stroke. Animal models of thrombosis have identified GPVI as a promising target for antithrombotic therapy. Although for many years the crystal structure of GPVI has been known, the essential details of its interaction with collagen have remained elusive. Here, we present crystal structures of the GPVI ectodomain bound to triple-helical collagen peptides, which reveal a collagen-binding site across the β-sheet of the D1 domain. Mutagenesis and binding studies confirm the observed binding site and identify Trp76, Arg38, and Glu40 as essential residues for binding to fibrillar collagens and collagen-related peptides (CRPs). GPVI binds a site on collagen comprising two collagen chains with the core formed by the sequence motif OGPOGP. Potent GPVI-binding peptides from Toolkit-III all contain OGPOGP; weaker binding peptides frequently contain a partial motif varying at either terminus. Alanine-scanning of peptide III-30 also identified two AGPOGP motifs that contribute to GPVI binding, but steric hindrance between GPVI molecules restricts the maximum binding capacity. We further show that no cooperative interactions could occur between two GPVI monomers binding to a stretch of (GPO)5 and that binding of ≥2 GPVI molecules to a fibril-embedded helix requires non-overlapping OGPOGP motifs. Our structure confirms the previously suggested similarity in collagen binding between GPVI and leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1) but also indicates significant differences that may be exploited for the development of receptor-specific therapeutics.
Collapse
|
12
|
Picker J, Lan Z, Arora S, Green M, Hahn M, Cosgriff-Hernandez E, Hook M. Prokaryotic Collagen-Like Proteins as Novel Biomaterials. Front Bioeng Biotechnol 2022; 10:840939. [PMID: 35372322 PMCID: PMC8968730 DOI: 10.3389/fbioe.2022.840939] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/10/2022] [Indexed: 12/13/2022] Open
Abstract
Collagens are the major structural component in animal extracellular matrices and are critical signaling molecules in various cell-matrix interactions. Its unique triple helical structure is enabled by tripeptide Gly-X-Y repeats. Understanding of sequence requirements for animal-derived collagen led to the discovery of prokaryotic collagen-like protein in the early 2000s. These prokaryotic collagen-like proteins are structurally similar to mammalian collagens in many ways. However, unlike the challenges associated with recombinant expression of mammalian collagens, these prokaryotic collagen-like proteins can be readily expressed in E. coli and are amenable to genetic modification. In this review article, we will first discuss the properties of mammalian collagen and provide a comparative analysis of mammalian collagen and prokaryotic collagen-like proteins. We will then review the use of prokaryotic collagen-like proteins to both study the biology of conventional collagen and develop a new biomaterial platform. Finally, we will describe the application of Scl2 protein, a streptococcal collagen-like protein, in thromboresistant coating for cardiovascular devices, scaffolds for bone regeneration, chronic wound dressing and matrices for cartilage regeneration.
Collapse
Affiliation(s)
- Jonathan Picker
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M, Houston, TX, United States
| | - Ziyang Lan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Srishtee Arora
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M, Houston, TX, United States
| | - Mykel Green
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Mariah Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| | | | - Magnus Hook
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M, Houston, TX, United States
| |
Collapse
|
13
|
Lepucki A, Orlińska K, Mielczarek-Palacz A, Kabut J, Olczyk P, Komosińska-Vassev K. The Role of Extracellular Matrix Proteins in Breast Cancer. J Clin Med 2022; 11:jcm11051250. [PMID: 35268340 PMCID: PMC8911242 DOI: 10.3390/jcm11051250] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/16/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix is a structure composed of many molecules, including fibrillar (types I, II, III, V, XI, XXIV, XXVII) and non-fibrillar collagens (mainly basement membrane collagens: types IV, VIII, X), non-collagenous glycoproteins (elastin, laminin, fibronectin, thrombospondin, tenascin, osteopontin, osteonectin, entactin, periostin) embedded in a gel of negatively charged water-retaining glycosaminoglycans (GAGs) such as non-sulfated hyaluronic acid (HA) and sulfated GAGs which are linked to a core protein to form proteoglycans (PGs). This highly dynamic molecular network provides critical biochemical and biomechanical cues that mediate the cell–cell and cell–matrix interactions, influence cell growth, migration and differentiation and serve as a reservoir of cytokines and growth factors’ action. The breakdown of normal ECM and its replacement with tumor ECM modulate the tumor microenvironment (TME) composition and is an essential part of tumorigenesis and metastasis, acting as key driver for malignant progression. Abnormal ECM also deregulate behavior of stromal cells as well as facilitating tumor-associated angiogenesis and inflammation. Thus, the tumor matrix modulates each of the classically defined hallmarks of cancer promoting the growth, survival and invasion of the cancer. Moreover, various ECM-derived components modulate the immune response affecting T cells, tumor-associated macrophages (TAM), dendritic cells and cancer-associated fibroblasts (CAF). This review article considers the role that extracellular matrix play in breast cancer. Determining the detailed connections between the ECM and cellular processes has helped to identify novel disease markers and therapeutic targets.
Collapse
Affiliation(s)
- Arkadiusz Lepucki
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.L.); (K.O.)
| | - Kinga Orlińska
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.L.); (K.O.)
| | - Aleksandra Mielczarek-Palacz
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.M.-P.); (J.K.)
| | - Jacek Kabut
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.M.-P.); (J.K.)
| | - Pawel Olczyk
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.L.); (K.O.)
- Correspondence:
| | - Katarzyna Komosińska-Vassev
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland;
| |
Collapse
|
14
|
Momin N, Palmeri JR, Lutz EA, Jailkhani N, Mak H, Tabet A, Chinn MM, Kang BH, Spanoudaki V, Hynes RO, Wittrup KD. Maximizing response to intratumoral immunotherapy in mice by tuning local retention. Nat Commun 2022; 13:109. [PMID: 35013154 PMCID: PMC8748612 DOI: 10.1038/s41467-021-27390-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 11/17/2021] [Indexed: 01/08/2023] Open
Abstract
Direct injection of therapies into tumors has emerged as an administration route capable of achieving high local drug exposure and strong anti-tumor response. A diverse array of immune agonists ranging in size and target are under development as local immunotherapies. However, due to the relatively recent adoption of intratumoral administration, the pharmacokinetics of locally-injected biologics remains poorly defined, limiting rational design of tumor-localized immunotherapies. Here we define a pharmacokinetic framework for biologics injected intratumorally that can predict tumor exposure and effectiveness. We find empirically and computationally that extending the tumor exposure of locally-injected interleukin-2 by increasing molecular size and/or improving matrix-targeting affinity improves therapeutic efficacy in mice. By tracking the distribution of intratumorally-injected proteins using positron emission tomography, we observe size-dependent enhancement in tumor exposure occurs by slowing the rate of diffusive escape from the tumor and by increasing partitioning to an apparent viscous region of the tumor. In elucidating how molecular weight and matrix binding interplay to determine tumor exposure, our model can aid in the design of intratumoral therapies to exert maximal therapeutic effect.
Collapse
Affiliation(s)
- Noor Momin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Joseph R Palmeri
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Emi A Lutz
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Noor Jailkhani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Howard Mak
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Anthony Tabet
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Magnolia M Chinn
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Byong H Kang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Virginia Spanoudaki
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Richard O Hynes
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - K Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA.
| |
Collapse
|
15
|
Karamanos NK, Theocharis AD, Piperigkou Z, Manou D, Passi A, Skandalis SS, Vynios DH, Orian-Rousseau V, Ricard-Blum S, Schmelzer CEH, Duca L, Durbeej M, Afratis NA, Troeberg L, Franchi M, Masola V, Onisto M. A guide to the composition and functions of the extracellular matrix. FEBS J 2021; 288:6850-6912. [PMID: 33605520 DOI: 10.1111/febs.15776] [Citation(s) in RCA: 482] [Impact Index Per Article: 120.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Extracellular matrix (ECM) is a dynamic 3-dimensional network of macromolecules that provides structural support for the cells and tissues. Accumulated knowledge clearly demonstrated over the last decade that ECM plays key regulatory roles since it orchestrates cell signaling, functions, properties and morphology. Extracellularly secreted as well as cell-bound factors are among the major members of the ECM family. Proteins/glycoproteins, such as collagens, elastin, laminins and tenascins, proteoglycans and glycosaminoglycans, hyaluronan, and their cell receptors such as CD44 and integrins, responsible for cell adhesion, comprise a well-organized functional network with significant roles in health and disease. On the other hand, enzymes such as matrix metalloproteinases and specific glycosidases including heparanase and hyaluronidases contribute to matrix remodeling and affect human health. Several cell processes and functions, among them cell proliferation and survival, migration, differentiation, autophagy, angiogenesis, and immunity regulation are affected by certain matrix components. Structural alterations have been also well associated with disease progression. This guide on the composition and functions of the ECM gives a broad overview of the matrisome, the major ECM macromolecules, and their interaction networks within the ECM and with the cell surface, summarizes their main structural features and their roles in tissue organization and cell functions, and emphasizes the importance of specific ECM constituents in disease development and progression as well as the advances in molecular targeting of ECM to design new therapeutic strategies.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Demitrios H Vynios
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Véronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems- Functional Molecular Systems, Eggenstein-Leopoldshafen, Germany
| | - Sylvie Ricard-Blum
- University of Lyon, UMR 5246, ICBMS, Université Lyon 1, CNRS, Villeurbanne Cedex, France
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany
- Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2: Matrix Aging and Vascular Remodelling, Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Madeleine Durbeej
- Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, Sweden
| | - Nikolaos A Afratis
- Department Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Linda Troeberg
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich, UK
| | - Marco Franchi
- Department for Life Quality Study, University of Bologna, Rimini, Italy
| | | | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
16
|
van der Wijst MGP, Vazquez SE, Hartoularos GC, Bastard P, Grant T, Bueno R, Lee DS, Greenland JR, Sun Y, Perez R, Ogorodnikov A, Ward A, Mann SA, Lynch KL, Yun C, Havlir DV, Chamie G, Marquez C, Greenhouse B, Lionakis MS, Norris PJ, Dumont LJ, Kelly K, Zhang P, Zhang Q, Gervais A, Le Voyer T, Whatley A, Si Y, Byrne A, Combes AJ, Rao AA, Song YS, Fragiadakis GK, Kangelaris K, Calfee CS, Erle DJ, Hendrickson C, Krummel MF, Woodruff PG, Langelier CR, Casanova JL, Derisi JL, Anderson MS, Ye CJ. Type I interferon autoantibodies are associated with systemic immune alterations in patients with COVID-19. Sci Transl Med 2021; 13:eabh2624. [PMID: 34429372 PMCID: PMC8601717 DOI: 10.1126/scitranslmed.abh2624] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/13/2021] [Indexed: 12/11/2022]
Abstract
Neutralizing autoantibodies against type I interferons (IFNs) have been found in some patients with critical coronavirus disease 2019 (COVID-19), the disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the prevalence of these antibodies, their longitudinal dynamics across the disease severity scale, and their functional effects on circulating leukocytes remain unknown. Here, in 284 patients with COVID-19, we found type I IFN–specific autoantibodies in peripheral blood samples from 19% of patients with critical disease and 6% of patients with severe disease. We found no type I IFN autoantibodies in individuals with moderate disease. Longitudinal profiling of over 600,000 peripheral blood mononuclear cells using multiplexed single-cell epitope and transcriptome sequencing from 54 patients with COVID-19 and 26 non–COVID-19 controls revealed a lack of type I IFN–stimulated gene (ISG-I) responses in myeloid cells from patients with critical disease. This was especially evident in dendritic cell populations isolated from patients with critical disease producing type I IFN–specific autoantibodies. Moreover, we found elevated expression of the inhibitory receptor leukocyte-associated immunoglobulin-like receptor 1 (LAIR1) on the surface of monocytes isolated from patients with critical disease early in the disease course. LAIR1 expression is inversely correlated with ISG-I expression response in patients with COVID-19 but is not expressed in healthy controls. The deficient ISG-I response observed in patients with critical COVID-19 with and without type I IFN–specific autoantibodies supports a unifying model for disease pathogenesis involving ISG-I suppression through convergent mechanisms.
Collapse
Affiliation(s)
- Monique G. P. van der Wijst
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713AV Groningen, Netherlands
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sara E. Vazquez
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94143, USA
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - George C. Hartoularos
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- University of Paris, Imagine Institute, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Tianna Grant
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Raymund Bueno
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David S. Lee
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - John R. Greenland
- Department of Medicine, University of California, San Francisco, San Francisco Medical Service, San Francisco Veterans Affairs Health Care System, San Francisco, CA 94121, USA
| | - Yang Sun
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Richard Perez
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Anton Ogorodnikov
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alyssa Ward
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sabrina A. Mann
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Kara L. Lynch
- Zuckerberg San Francisco General, San Francisco, CA 94110, USA
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Cassandra Yun
- Zuckerberg San Francisco General, San Francisco, CA 94110, USA
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Diane V. Havlir
- Division of HIV, Infectious Disease and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gabriel Chamie
- Division of HIV, Infectious Disease and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Carina Marquez
- Division of HIV, Infectious Disease and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bryan Greenhouse
- Division of HIV, Infectious Disease and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michail S. Lionakis
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Philip J. Norris
- Zuckerberg San Francisco General, San Francisco, CA 94110, USA
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Vitalant Research Institute, San Francisco, CA 94118, USA
| | - Larry J. Dumont
- Vitalant Research Institute, Denver, CO 80230, USA
- University of Colorado School of Medicine, Aurora, CO 80045, USA
- Geisel School of Medicine at Dartmouth, Lebanon, NH 03755, USA
| | | | - Peng Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Qian Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Adrian Gervais
- University of Paris, Imagine Institute, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Tom Le Voyer
- University of Paris, Imagine Institute, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Alexander Whatley
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yichen Si
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ashley Byrne
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Alexis J. Combes
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arjun Arkal Rao
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yun S. Song
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Statistics, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gabriela K. Fragiadakis
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kirsten Kangelaris
- Division of Hospital Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Carolyn S. Calfee
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine and the Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David J. Erle
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
- Zuckerberg San Francisco General, San Francisco, CA 94110, USA
| | - Carolyn Hendrickson
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine and the Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew F. Krummel
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Prescott G. Woodruff
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine and the Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Charles R. Langelier
- Division of Infectious Disease, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- University of Paris, Imagine Institute, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Joseph L. Derisi
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Mark S. Anderson
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Chun Jimmie Ye
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Departments of Epidemiology and Biostatistics and Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - on behalf of the UCSF COMET consortium
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713AV Groningen, Netherlands
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94143, USA
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France
- University of Paris, Imagine Institute, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Medicine, University of California, San Francisco, San Francisco Medical Service, San Francisco Veterans Affairs Health Care System, San Francisco, CA 94121, USA
- School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Zuckerberg San Francisco General, San Francisco, CA 94110, USA
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of HIV, Infectious Disease and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20814, USA
- Vitalant Research Institute, San Francisco, CA 94118, USA
- Vitalant Research Institute, Denver, CO 80230, USA
- University of Colorado School of Medicine, Aurora, CO 80045, USA
- Geisel School of Medicine at Dartmouth, Lebanon, NH 03755, USA
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Statistics, University of California, Berkeley, Berkeley, CA 94720, USA
- Division of Hospital Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine and the Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Infectious Disease, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Departments of Epidemiology and Biostatistics and Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| |
Collapse
|
17
|
Xie Y, Li X, Chai Y, Song H, Qi J, Gao GF. Structural basis of malarial parasite RIFIN-mediated immune escape against LAIR1. Cell Rep 2021; 36:109600. [PMID: 34433057 DOI: 10.1016/j.celrep.2021.109600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/17/2021] [Accepted: 08/03/2021] [Indexed: 11/16/2022] Open
Abstract
Malaria infection by Plasmodium falciparum continues to pose a global threat to the human population. P. falciparum expresses variable erythrocyte surface antigens such as RIFINs. Public antibodies with LAIR1 insertion have been identified from malarial patients against a subset of RIFINs. In this study, we solve a LAIR1-binding RIFIN structure: the complex structures of two RIFINs bound to mutated or wild-type LAIR1 in two distinct patterns. Notably, the two RIFINs engage similar binding sites on LAIR1 with different angles, and the RIFIN-binding sites overlap with the collagen-binding site. Surprisingly, RIFINs use completely different binding sites to bind to LAIR1 or LILRB1, indicating the kaleidoscopic change of RIFINs. We then verify that RIFIN could induce LAIR1-mediated cell signaling, and LAIR1-containing antibodies could block the pathway. The findings of this study provide structural insights into the mechanism of the immune escape of P. falciparum and the endless arms race between parasite and host.
Collapse
Affiliation(s)
- Yijia Xie
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Li
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hao Song
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Jianxun Qi
- University of Chinese Academy of Sciences, Beijing 100049, China; CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - George F Gao
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Influenza Research and Early Warning (CASCIRE), Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
18
|
Xu K, Wang Y, Shen CH, Chen Y, Zhang B, Liu K, Tsybovsky Y, Wang S, Farney SK, Gorman J, Stephens T, Verardi R, Yang Y, Zhou T, Chuang GY, Lanzavecchia A, Piccoli L, Kwong PD. Structural basis of LAIR1 targeting by polymorphic Plasmodium RIFINs. Nat Commun 2021; 12:4226. [PMID: 34244481 PMCID: PMC8270905 DOI: 10.1038/s41467-021-24291-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/11/2021] [Indexed: 11/18/2022] Open
Abstract
RIFIN, a large family of Plasmodium variant surface antigens, plays a crucial role in malaria pathogenesis by mediating immune suppression through activation of inhibitory receptors such as LAIR1, and antibodies with LAIR1 inserts have been identified that bind infected erythrocytes through RIFIN. However, details of RIFIN-mediated LAIR1 recognition and receptor activation have been unclear. Here, we use negative-stain EM to define the architecture of LAIR1-inserted antibodies and determine crystal structures of RIFIN-variable 2 (V2) domain in complex with a LAIR1 domain. These structures reveal the LAIR1-binding region of RIFIN to be hydrophobic and membrane-distal, to exhibit extensive structural diversity, and to interact with RIFIN-V2 in a one-to-one fashion. Through structural and sequence analysis of various LAIR1 constructs, we identify essential elements of RIFIN-binding on LAIR1. Furthermore, a structure-derived LAIR1-binding sequence signature ascertained >20 LAIR1-binding RIFINs, including some from P. falciparum field strains and Plasmodium species infecting gorillas and chimpanzees.
Collapse
MESH Headings
- Antibodies, Protozoan/genetics
- Antibodies, Protozoan/metabolism
- Antigenic Variation/genetics
- Antigens, Protozoan/immunology
- Antigens, Protozoan/metabolism
- Antigens, Protozoan/ultrastructure
- Crystallography, X-Ray
- Humans
- Malaria, Falciparum/immunology
- Malaria, Falciparum/parasitology
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Membrane Proteins/ultrastructure
- Mutation
- Plasmodium falciparum/immunology
- Plasmodium falciparum/metabolism
- Protein Domains/genetics
- Protozoan Proteins/immunology
- Protozoan Proteins/metabolism
- Protozoan Proteins/ultrastructure
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/ultrastructure
Collapse
Affiliation(s)
- Kai Xu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.
| | - Yiran Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chen-Hsiang Shen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yiwei Chen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, ETH Zurich, Wolfgang-Pauli-Strasse 10, Zurich, Switzerland
| | - Baoshan Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kevin Liu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yaroslav Tsybovsky
- Electron Microscopy Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Shuishu Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - S Katie Farney
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jason Gorman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tyler Stephens
- Electron Microscopy Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Raffaello Verardi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yongping Yang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Gwo-Yu Chuang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- National Institute of Molecular Genetics (INGM), Milano, Italy
| | - Luca Piccoli
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
19
|
Keerthivasan S, Şenbabaoğlu Y, Martinez-Martin N, Husain B, Verschueren E, Wong A, Yang YA, Sun Y, Pham V, Hinkle T, Oei Y, Madireddi S, Corpuz R, Tam L, Carlisle S, Roose-Girma M, Modrusan Z, Ye Z, Koerber JT, Turley SJ. Homeostatic functions of monocytes and interstitial lung macrophages are regulated via collagen domain-binding receptor LAIR1. Immunity 2021; 54:1511-1526.e8. [PMID: 34260887 DOI: 10.1016/j.immuni.2021.06.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 02/21/2021] [Accepted: 06/14/2021] [Indexed: 12/18/2022]
Abstract
Myeloid cells encounter stromal cells and their matrix determinants on a continual basis during their residence in any given organ. Here, we examined the impact of the collagen receptor LAIR1 on myeloid cell homeostasis and function. LAIR1 was highly expressed in the myeloid lineage and enriched in non-classical monocytes. Proteomic definition of the LAIR1 interactome identified stromal factor Colec12 as a high-affinity LAIR1 ligand. Proteomic profiling of LAIR1 signaling triggered by Collagen1 and Colec12 highlighted pathways associated with survival, proliferation, and differentiation. Lair1-/- mice had reduced frequencies of Ly6C- monocytes, which were associated with altered proliferation and apoptosis of non-classical monocytes from bone marrow and altered heterogeneity of interstitial macrophages in lung. Myeloid-specific LAIR1 deficiency promoted metastatic growth in a melanoma model and LAIR1 expression associated with improved clinical outcomes in human metastatic melanoma. Thus, monocytes and macrophages rely on LAIR1 sensing of stromal determinants for fitness and function, with relevance in homeostasis and disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Anne Wong
- Genentech Inc., South San Francisco, CA, USA
| | | | | | | | | | - Yoko Oei
- Genentech Inc., South San Francisco, CA, USA
| | | | | | - Lucinda Tam
- Genentech Inc., South San Francisco, CA, USA
| | | | | | | | - Zhengmao Ye
- Genentech Inc., South San Francisco, CA, USA
| | | | | |
Collapse
|
20
|
Of myeloid cells and fibroblasts-A love story. Immunity 2021; 54:1371-1373. [PMID: 34260884 DOI: 10.1016/j.immuni.2021.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The interaction between myeloid cells and the extracellular matrix is important for tissue homeostasis and pathophysiology. In this issue of Immunity, Keerthivasan et al. reveal crosstalk dependent on the collagen receptor LAIR1 that regulates the dynamics of monocytes and macrophages during steady-state and cancer.
Collapse
|
21
|
Molecular Basis of Complement C1q Collagen-Like Region Interaction with the Immunoglobulin-Like Receptor LAIR-1. Int J Mol Sci 2021; 22:ijms22105125. [PMID: 34066122 PMCID: PMC8151509 DOI: 10.3390/ijms22105125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/16/2021] [Accepted: 05/05/2021] [Indexed: 02/08/2023] Open
Abstract
The immune system homeostasis relies on a tight equilibrium of interconnected stimulatory and inhibitory signals. Disruption of this balance is characteristic of autoimmune diseases such as systemic lupus erythematosus (SLE). Aside from activating the classical complement pathway and enhancing pathogens and apoptotic cells phagocytosis, C1q has been recently shown to play an important role in immune modulation and tolerance by interacting with several inhibitory and stimulatory immune receptors. Due to its functional organization into collagen-like (CLR) and globular (GR) regions and its multimeric nature, C1q is able to interact simultaneously with several of these receptors and locally congregate pro- and anti-inflammatory signals, thus modulating the immune response. Leukocyte associated immunoglobulin-like (Ig-like) receptor 1 (LAIR-1), a ubiquitous collagen receptor expressed in many immune cell types, has been reported to interact with the CLR of C1q. In this study, we provide new insights into the molecular and structural determinants underlying C1q/LAIR-1 interaction. Recombinant LAIR-1 extracellular Ig-like domain was produced and tested for its interaction with C1q. A molecular dissection of C1q combined with competition assays reveals that LAIR-1 interacts with C1q’s CLR through a binding site close but different from the one of its associated C1r2s2 proteases tetramer. On the other side, we identified LAIR-1 residues involved in C1q interaction by site-directed mutational analysis. All together, these results lead to propose a possible model for C1q interaction with LAIR-1 and will contribute to the fundamental understanding of C1q-mediated immune tolerance.
Collapse
|
22
|
Nedeva IR, Vitale M, Elson A, Hoyland JA, Bella J. Role of OSCAR Signaling in Osteoclastogenesis and Bone Disease. Front Cell Dev Biol 2021; 9:641162. [PMID: 33912557 PMCID: PMC8072347 DOI: 10.3389/fcell.2021.641162] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 03/15/2021] [Indexed: 11/13/2022] Open
Abstract
Formation of mature bone-resorbing cells through osteoclastogenesis is required for the continuous remodeling and repair of bone tissue. In aging and disease this process may become aberrant, resulting in excessive bone degradation and fragility fractures. Interaction of receptor-activator of nuclear factor-κB (RANK) with its ligand RANKL activates the main signaling pathway for osteoclastogenesis. However, compelling evidence indicates that this pathway may not be sufficient for the production of mature osteoclast cells and that co-stimulatory signals may be required for both the expression of osteoclast-specific genes and the activation of osteoclasts. Osteoclast-associated receptor (OSCAR), a regulator of osteoclast differentiation, provides one such co-stimulatory pathway. This review summarizes our present knowledge of osteoclastogenesis signaling and the role of OSCAR in the normal production of bone-resorbing cells and in bone disease. Understanding the signaling mechanism through this receptor and how it contributes to the production of mature osteoclasts may offer a more specific and targeted approach for pharmacological intervention against pathological bone resorption.
Collapse
Affiliation(s)
- Iva R Nedeva
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Mattia Vitale
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Judith A Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Jordi Bella
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
23
|
Damaskinaki FN, Moran LA, Garcia A, Kellam B, Watson SP. Overcoming challenges in developing small molecule inhibitors for GPVI and CLEC-2. Platelets 2021; 32:744-752. [PMID: 33406951 DOI: 10.1080/09537104.2020.1863939] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
GPVI and CLEC-2 have emerged as promising targets for long-term prevention of both arterial thrombosis and thrombo-inflammation with a decreased bleeding risk relative to current drugs. However, while there are potent blocking antibodies of both receptors, their protein nature comes with decreased bioavailability, making formulation for oral medication challenging. Small molecules are able to overcome these limitations, but there are many challenges in developing antagonists of nanomolar potency, which is necessary when considering the structural features that underlie the interaction of CLEC-2 and GPVI with their protein ligands. In this review, we describe current small-molecule inhibitors for both receptors and strategies to overcome such limitations, including considerations when it comes to in silico drug design and the importance of complex compound library selection.
Collapse
Affiliation(s)
- Foteini-Nafsika Damaskinaki
- Institute of Cardiovascular Sciences, Level 1 IBR, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Birmingham and Nottingham, The Midlands, UK.,Biodiscovery Institute, University Park, University of Nottingham, Nottingham, UK
| | - Luis A Moran
- Institute of Cardiovascular Sciences, Level 1 IBR, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, and Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Angel Garcia
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, and Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Barrie Kellam
- Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Birmingham and Nottingham, The Midlands, UK.,Biodiscovery Institute, University Park, University of Nottingham, Nottingham, UK
| | - Steve P Watson
- Institute of Cardiovascular Sciences, Level 1 IBR, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Birmingham and Nottingham, The Midlands, UK
| |
Collapse
|
24
|
Jürgensen HJ, van Putten S, Nørregaard KS, Bugge TH, Engelholm LH, Behrendt N, Madsen DH. Cellular uptake of collagens and implications for immune cell regulation in disease. Cell Mol Life Sci 2020; 77:3161-3176. [PMID: 32100084 PMCID: PMC11105017 DOI: 10.1007/s00018-020-03481-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/03/2020] [Accepted: 02/07/2020] [Indexed: 12/15/2022]
Abstract
As the dominant constituent of the extracellular matrix (ECM), collagens of different types are critical for the structural properties of tissues and make up scaffolds for cellular adhesion and migration. Importantly, collagens also directly modulate the phenotypic state of cells by transmitting signals that influence proliferation, differentiation, polarization, survival, and more, to cells of mesenchymal, epithelial, or endothelial origin. Recently, the potential of collagens to provide immune regulatory signals has also been demonstrated, and it is believed that pathological changes in the ECM shape immune cell phenotype. Collagens are themselves heavily regulated by a multitude of structural modulations or by catabolic pathways. One of these pathways involves a cellular uptake of collagens or soluble collagen-like defense collagens of the innate immune system mediated by endocytic collagen receptors. This cellular uptake is followed by the degradation of collagens in lysosomes. The potential of this pathway to regulate collagens in pathological conditions is evident from the increased extracellular accumulation of both collagens and collagen-like defense collagens following endocytic collagen receptor ablation. Here, we review how endocytic collagen receptors regulate collagen turnover during physiological conditions and in pathological conditions, such as fibrosis and cancer. Furthermore, we highlight the potential of collagens to regulate immune cells and discuss how endocytic collagen receptors can directly regulate immune cell activity in pathological conditions or do it indirectly by altering the extracellular milieu. Finally, we discuss the potential collagen receptors utilized by immune cells to directly detect ECM-related changes in the tissues which they encounter.
Collapse
Affiliation(s)
- Henrik J Jürgensen
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center, University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen N, Denmark.
| | - Sander van Putten
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center, University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen N, Denmark
| | - Kirstine S Nørregaard
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center, University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen N, Denmark
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lars H Engelholm
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center, University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen N, Denmark
| | - Niels Behrendt
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center, University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen N, Denmark
| | - Daniel H Madsen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730, Herlev, Denmark.
| |
Collapse
|
25
|
Rowley AT, Meli VS, Wu-Woods NJ, Chen EY, Liu WF, Wang SW. Effects of Surface-Bound Collagen-Mimetic Peptides on Macrophage Uptake and Immunomodulation. Front Bioeng Biotechnol 2020; 8:747. [PMID: 32719788 PMCID: PMC7348040 DOI: 10.3389/fbioe.2020.00747] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/11/2020] [Indexed: 11/13/2022] Open
Abstract
The interaction between collagen/collagen-like peptides and the commonly expressed immune cell receptor LAIR-1 (leukocyte-associated immunoglobulin-like receptor-1) regulates and directs immune responses throughout the body. Understanding and designing these interactions within the context of biomaterials could advance the development of materials used in medical applications. In this study, we investigate the immunomodulatory effects of biomaterials engineered to display a human collagen III-derived ligand peptide (LAIR1-LP) that targets LAIR-1. Specifically, we examine the effects of LAIR1-LP functionalized surfaces on uptake of polymeric particles and cell debris by macrophages polarized toward inflammatory or healing phenotypes. We observed that culture of macrophages on LAIR1-LP functionalized surfaces increased their uptake of PLGA micro- and nano-particles, as well as apoptotic fibroblasts, while reducing their secretion of TNFα in response to LPS/IFNγ pro-inflammatory stimulation, when compared to cells seeded on control surfaces. To investigate the role of LAIR-1 in the observed LAIR1-LP-induced effects, we used siRNA to knock down LAIR-1 expression and found that cells lacking LAIR-1 exhibited enhanced particle uptake on LAIR1-LP and control surfaces. Furthermore, analysis of gene expression showed that LAIR-1 knockdown led to increase expression of other receptors involved in cell uptake, including CD-36, SRA-1, and beta-2 integrin. Together, our study suggests that LAIR1-LP enhances macrophage uptake potentially through interactions with collagen-domain binding surface receptors, and inhibits inflammation through interaction with LAIR-1.
Collapse
Affiliation(s)
- Andrew T Rowley
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, CA, United States
| | - Vijaykumar S Meli
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, CA, United States.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | - Natalie J Wu-Woods
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, CA, United States
| | - Esther Y Chen
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, CA, United States
| | - Wendy F Liu
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, CA, United States.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States.,The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, United States
| | - Szu-Wen Wang
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, CA, United States.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
26
|
Hoop CL, Zhu J, Bhattacharya S, Tobita CA, Radford SE, Baum J. Collagen I Weakly Interacts with the β-Sheets of β 2-Microglobulin and Enhances Conformational Exchange To Induce Amyloid Formation. J Am Chem Soc 2020; 142:1321-1331. [PMID: 31875390 PMCID: PMC7135851 DOI: 10.1021/jacs.9b10421] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
![]()
Amyloidogenesis is
significant in both protein function and pathology.
Amyloid formation of folded, globular proteins is commonly initiated
by partial or complete unfolding. However, how this unfolding event
is triggered for proteins that are otherwise stable in their native
environments is not well understood. The accumulation of the immunoglobulin
protein β2-microglobulin (β2m) into
amyloid plaques in the joints of long-term hemodialysis patients is
the hallmark of dialysis-related amyloidosis (DRA). While β2m does not form amyloid unassisted near neutral pH in vitro, the localization of β2m deposits
to joint spaces suggests a role for the local extracellular matrix
(ECM) proteins, specifically collagens, in promoting amyloid formation.
Indeed, collagen and other ECM components have been observed to facilitate
β2m amyloid formation, but the large size and anisotropy
of the complex, combined with the low affinity of these interactions,
have limited atomic-level elucidation of the amyloid-promoting mechanism(s)
by these molecules. Using solution NMR approaches that uniquely probe
weak interactions in large molecular weight complexes, we are able
to map the binding interfaces on β2m for collagen
I and detect collagen I-induced μs–ms time-scale dynamics
in the β2m backbone. By combining solution NMR relaxation
methods and 15N-dark-state exchange saturation transfer
experiments, we propose a model in which weak, multimodal collagen
I−β2m interactions promote exchange with a
minor population of amyloid-competent species to induce fibrillogenesis.
The results portray the intimate role of the environment in switching
an innocuous protein into an amyloid-competent state, rationalizing
the localization of amyloid deposits in DRA.
Collapse
Affiliation(s)
- Cody L Hoop
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
| | - Jie Zhu
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
| | | | - Caitlyn A Tobita
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, Faculty of Biological Sciences , University of Leeds , Leeds LS2 9JT , U.K
| | - Jean Baum
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
| |
Collapse
|
27
|
Barrow AD, Martin CJ, Colonna M. The Natural Cytotoxicity Receptors in Health and Disease. Front Immunol 2019; 10:909. [PMID: 31134055 PMCID: PMC6514059 DOI: 10.3389/fimmu.2019.00909] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/09/2019] [Indexed: 12/31/2022] Open
Abstract
The Natural Cytotoxicity Receptors (NCRs), NKp46, NKp44, and NKp30, were some of the first human activating Natural Killer (NK) cell receptors involved in the non-MHC-restricted recognition of tumor cells to be cloned over 20 years ago. Since this time many host- and pathogen-encoded ligands have been proposed to bind the NCRs and regulate the cytotoxic and cytokine-secreting functions of tissue NK cells. This diverse set of NCR ligands can manifest on the surface of tumor or virus-infected cells or can be secreted extracellularly, suggesting a remarkable NCR polyfunctionality that regulates the activity of NK cells in different tissue compartments during steady state or inflammation. Moreover, the NCRs can also be expressed by other innate and adaptive immune cell subsets under certain tissue conditions potentially conferring NK recognition programs to these cells. Here we review NCR biology in health and disease with particular reference to how this important class of receptors regulates the functions of tissue NK cells as well as confer NK cell recognition patterns to other innate and adaptive lymphocyte subsets. Finally, we highlight how NCR biology is being harnessed for novel therapeutic interventions particularly for enhanced tumor surveillance.
Collapse
Affiliation(s)
- Alexander David Barrow
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Claudia Jane Martin
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
28
|
Biassoni R, Malnati MS. Human Natural Killer Receptors, Co-Receptors, and Their Ligands. ACTA ACUST UNITED AC 2019; 121:e47. [PMID: 30040219 DOI: 10.1002/cpim.47] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the last 20 years, the study of human natural killer (NK) cells has moved from the first molecular characterizations of very few receptor molecules to the identification of a plethora of receptors displaying surprisingly divergent functions. We have contributed to the description of inhibitory receptors and their signaling pathways, important in fine regulation in many cell types, but unknown until their discovery in the NK cells. Inhibitory function is central to regulating NK-mediated cytolysis, with different molecular structures evolving during speciation to assure its persistence. More recently, it has become possible to characterize the NK triggering receptors mediating natural cytotoxicity, unveiling the existence of a network of cellular interactions between effectors of both natural and adaptive immunity. This unit reviews the contemporary history of molecular studies of receptors and ligands involved in NK cell function, characterizing the ligands of the triggering receptor and the mechanisms for finely regulating their expression in pathogen-infected or tumor cells. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Roberto Biassoni
- IRCCS Istituto Giannina Gaslini, Laboratory of Molecular Medicine, Genova, Italy
| | - Mauro S Malnati
- IRCCS Ospedale San Raffaele, Unit of Human Virology, Division of Immunology, Transplantation and Infectious Diseases, Milan, Italy
| |
Collapse
|
29
|
Wu X, Zhang L, Zhou J, Liu L, Fu Q, Fu A, Feng X, Xin R, Liu H, Gao Y, Xue J. Clinicopathologic significance of LAIR-1 expression in hepatocellular carcinoma. Curr Probl Cancer 2019; 43:18-26. [DOI: 10.1016/j.currproblcancer.2018.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 04/04/2018] [Accepted: 04/20/2018] [Indexed: 12/22/2022]
|
30
|
Grover SP, Bergmeier W, Mackman N. Platelet Signaling Pathways and New Inhibitors. Arterioscler Thromb Vasc Biol 2019; 38:e28-e35. [PMID: 29563117 DOI: 10.1161/atvbaha.118.310224] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Steven P Grover
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine (S.P.G., N.M.) and McAllister Heart Institute and Department of Biochemistry and Biophysics (W.B.), University of North Carolina at Chapel Hill
| | - Wolfgang Bergmeier
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine (S.P.G., N.M.) and McAllister Heart Institute and Department of Biochemistry and Biophysics (W.B.), University of North Carolina at Chapel Hill
| | - Nigel Mackman
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine (S.P.G., N.M.) and McAllister Heart Institute and Department of Biochemistry and Biophysics (W.B.), University of North Carolina at Chapel Hill.
| |
Collapse
|
31
|
|
32
|
Slater A, Perrella G, Onselaer MB, Martin EM, Gauer JS, Xu RG, Heemskerk JWM, Ariëns RAS, Watson SP. Does fibrin(ogen) bind to monomeric or dimeric GPVI, or not at all? Platelets 2018; 30:281-289. [DOI: 10.1080/09537104.2018.1508649] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Gina Perrella
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| | - Marie-Blanche Onselaer
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Eleyna M Martin
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Julia S Gauer
- Thrombosis and Tissue Repair Group, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Rui-Gang Xu
- Thrombosis and Tissue Repair Group, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Johan WM Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| | - Robert A S Ariëns
- Thrombosis and Tissue Repair Group, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
33
|
Jin J, Wang Y, Ma Q, Wang N, Guo W, Jin B, Fang L, Chen L. LAIR-1 activation inhibits inflammatory macrophage phenotype in vitro. Cell Immunol 2018; 331:78-84. [PMID: 29887420 DOI: 10.1016/j.cellimm.2018.05.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 04/25/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022]
Abstract
Macrophages are key cell types of innate immunity and play a central role in inflammation and host defense. Leukocyte-associated Ig-like receptor-1 (LAIR-1) is highly expressed on macrophages and regulates macrophage functions in several conditions. However, whether LAIR-1 is involved in governing macrophage polarization is still not clear. Here, we investigated the effect of LAIR-1 on macrophage polarization using human macrophage polarization model with THP-1 cells. It was found that LAIR-1 was highly expressed in THP-1 macrophages. IFN-γ reduced LAIR-1 expression in THP-1 macrophages. However, IL-4 did not have an effect on the expression of LAIR-1. Moreover, activation of LAIR-1 significantly inhibited the proinflammatory M1-like macrophage differentiation and promoted alternative activation of macrophages. Therefore, LAIR-1 may play critical roles in macrophage polarization. This study provides a rationale for macrophage polarization and sheds light on homeostatic mechanism in which LAIR-1 activation can terminate inflammation which may be impaired in patients with autoimmune disease.
Collapse
Affiliation(s)
- Jingyi Jin
- Department of Immunology, The Fourth Military Medical University, Xi'an 710032, China
| | - Ying Wang
- Department of Stomatology, 307 Hospital, PLA, Beijing 100071, China
| | - Qianli Ma
- Department of Immunology, The Fourth Military Medical University, Xi'an 710032, China
| | - Ning Wang
- Department of Immunology, The Fourth Military Medical University, Xi'an 710032, China
| | - Wenwei Guo
- Department of Immunology, The Fourth Military Medical University, Xi'an 710032, China
| | - Boquan Jin
- Department of Immunology, The Fourth Military Medical University, Xi'an 710032, China
| | - Liang Fang
- Department of Immunology, The Fourth Military Medical University, Xi'an 710032, China.
| | - Lihua Chen
- Department of Immunology, The Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
34
|
Kim YK, Chu SH, Hsieh JY, Kamoku CM, Tenner AJ, Liu WF, Wang SW. Incorporation of a Ligand Peptide for Immune Inhibitory Receptor LAIR-1 on Biomaterial Surfaces Inhibits Macrophage Inflammatory Responses. Adv Healthc Mater 2017; 6. [PMID: 29083540 DOI: 10.1002/adhm.201700707] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/22/2017] [Indexed: 01/22/2023]
Abstract
Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) is an inhibitory receptor broadly expressed on immune cells, with its ligands residing within the extracellular matrix protein collagen. In this study, surfaces are modified with a LAIR-1 ligand peptide (LP), and it is observed that macrophages cultured on LAIR-1 LP-conjugated surfaces exhibit significantly reduced secretion of inflammatory cytokines in response to proinflammatory stimuli that reflect an injured environment. These downregulated mediators include TNF-α, MIP-1α, MIP-1β, MIP-2, RANTES, and MIG. Knockdown of LAIR-1 using siRNA abrogates this inhibition of cytokine secretion, supporting the specificity of the inhibitory effect to this receptor. These results are the first to demonstrate that integration of LAIR-1 ligands with biomaterials could suppress inflammatory responses.
Collapse
Affiliation(s)
- Yoon Kyung Kim
- Department of Chemical Engineering & Materials Science; University of California; Irvine CA 92697 USA
- Department of Biomedical Engineering; University of California; Irvine CA 92697 USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology; University of California; Irvine CA 92697 USA
| | - Shu-Hui Chu
- Department of Molecular Biology and Biochemistry; University of California; Irvine CA 92697 USA
| | - Jessica Y. Hsieh
- Department of Biomedical Engineering; University of California; Irvine CA 92697 USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology; University of California; Irvine CA 92697 USA
| | - Cody M. Kamoku
- Department of Chemical Engineering & Materials Science; University of California; Irvine CA 92697 USA
| | - Andrea J. Tenner
- Department of Molecular Biology and Biochemistry; University of California; Irvine CA 92697 USA
| | - Wendy F. Liu
- Department of Chemical Engineering & Materials Science; University of California; Irvine CA 92697 USA
- Department of Biomedical Engineering; University of California; Irvine CA 92697 USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology; University of California; Irvine CA 92697 USA
| | - Szu-Wen Wang
- Department of Chemical Engineering & Materials Science; University of California; Irvine CA 92697 USA
- Department of Biomedical Engineering; University of California; Irvine CA 92697 USA
| |
Collapse
|
35
|
Hoop CL, Zhu J, Nunes AM, Case DA, Baum J. Revealing Accessibility of Cryptic Protein Binding Sites within the Functional Collagen Fibril. Biomolecules 2017; 7:biom7040076. [PMID: 29104255 PMCID: PMC5745458 DOI: 10.3390/biom7040076] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/23/2017] [Accepted: 10/27/2017] [Indexed: 11/16/2022] Open
Abstract
Fibrillar collagens are the most abundant proteins in the extracellular matrix. Not only do they provide structural integrity to all of the connective tissues in the human body, but also their interactions with multiple cell receptors and other matrix molecules are essential to cell functions, such as growth, repair, and cell adhesion. Although specific binding sequences of several receptors have been determined along the collagen monomer, processes by which collagen binding partners recognize their binding sites in the collagen fibril, and the critical driving interactions, are poorly understood. The complex molecular assembly of bundled triple helices within the collagen fibril makes essential ligand binding sites cryptic or hidden from the molecular surface. Yet, critical biological processes that require collagen ligands to have access to interaction sites still occur. In this contribution, we will discuss the molecular packing of the collagen I fibril from the perspective of how collagen ligands access their known binding regions within the fibril, and we will present our analysis of binding site accessibility from the fibril surface. Understanding the basis of these interactions at the atomic level sets the stage for developing drug targets against debilitating collagen diseases and using collagen as drug delivery systems and new biomaterials.
Collapse
Affiliation(s)
- Cody L Hoop
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA.
| | - Jie Zhu
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA.
| | - Ana Monica Nunes
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA.
| | - David A Case
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA.
| | - Jean Baum
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
36
|
Abstract
Fibrin has recently been shown to activate platelets through the immunoglobulin receptor glycoprotein VI (GPVI). In the present study, we show that spreading of human platelets on fibrin is abolished in patients deficient in GPVI, confirming that fibrin activates human platelets through the immunoglobulin receptor. Using a series of proteolytic fragments, we show that D-dimer, but not the E fragment of fibrin, binds to GPVI and that immobilized D-dimer induces platelet spreading through activation of Src and Syk tyrosine kinases. In contrast, when platelets are activated in suspension, soluble D-dimer inhibits platelet aggregation induced by fibrin and collagen, but not by a collagen-related peptide composed of a repeat GPO sequence or by thrombin. Using surface plasmon resonance, we demonstrate that fibrin binds selectively to monomeric GPVI with a KD of 302 nM, in contrast to collagen, which binds primarily to dimeric GPVI. These results establish GPVI as the major signaling receptor for fibrin in human platelets and provide evidence that fibrin binds to a distinct configuration of GPVI. This indicates that it may be possible to develop agents that selectively block the interaction of fibrin but not collagen with the immunoglobulin receptor. Such agents are required to establish whether selective targeting of either interaction has the potential to lead to development of an antithrombotic agent with a reduced effect on bleeding relative to current antiplatelet drugs.
Collapse
|
37
|
Hsieh FL, Higgins MK. The structure of a LAIR1-containing human antibody reveals a novel mechanism of antigen recognition. eLife 2017; 6. [PMID: 28527239 PMCID: PMC5459573 DOI: 10.7554/elife.27311] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/08/2017] [Indexed: 01/11/2023] Open
Abstract
Antibodies are critical components of the human adaptive immune system, providing versatile scaffolds to display diverse antigen-binding surfaces. Nevertheless, most antibodies have similar architectures, with the variable immunoglobulin domains of the heavy and light chain each providing three hypervariable loops, which are varied to generate diversity. The recent identification of a novel class of antibody in humans from malaria endemic regions of Africa was therefore surprising as one hypervariable loop contains the entire collagen-binding domain of human LAIR1. Here, we present the structure of the Fab fragment of such an antibody. We show that its antigen-binding site has adopted an architecture that positions LAIR1, while itself being occluded. This therefore represents a novel means of antigen recognition, in which the Fab fragment of an antibody acts as an adaptor, linking a human protein insert with antigen-binding potential to the constant antibody regions which mediate immune cell recruitment. DOI:http://dx.doi.org/10.7554/eLife.27311.001 When bacteria, viruses or parasites invade the human body, the immune system responds by producing proteins called antibodies. Antibodies recognize and bind to molecules (known as antigens) on the surface of the invaders. This binding can either neutralize the invader directly or trigger signals that cause other parts of the immune system to destroy it. Our blood contains a huge range of different antibody molecules that each bind to a different antigen. This is despite most human antibodies having the same basic shape and structure. Six loops, known as complementarity determining regions (CDRs), emerge from the surface of the antibody to form the surface that recognizes the antigen. However, variations in the structure of the loops alter this surface enough to allow different antibodies to recognize completely different molecules. In 2016, a new class of antibodies was identified. Unlike previously identified antibodies, these molecules had an entire human protein, called LAIR1, inserted into one of their CDR loops. Members of this group of antibodies bind to a molecule, known as a RIFIN, that is found on the surface of human red blood cells that are infected with the parasite that causes malaria. How do LAIR1-containing antibodies bind to their RIFIN targets? Hsieh and Higgins investigated this question by using a technique called X-ray crystallography to determine the structure of the antibody. This revealed that instead of binding directly to an antigen, all of the six CDR loops in the LAIR1-containing antibody bind to the LAIR1 insert. By doing so, LAIR1 is oriented in a manner that enables it to bind to the RIFIN molecule from the parasite. This is the first known example of an antibody that recruits another protein to bind to an antigen rather than binding directly to the pathogen itself. A future challenge will be to see if other antibodies exist that use this mechanism and whether it can be employed to design new therapeutic antibodies. DOI:http://dx.doi.org/10.7554/eLife.27311.002
Collapse
Affiliation(s)
- Fu-Lien Hsieh
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Matthew K Higgins
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
38
|
Smith CW, Thomas SG, Raslan Z, Patel P, Byrne M, Lordkipanidzé M, Bem D, Meyaard L, Senis YA, Watson SP, Mazharian A. Mice Lacking the Inhibitory Collagen Receptor LAIR-1 Exhibit a Mild Thrombocytosis and Hyperactive Platelets. Arterioscler Thromb Vasc Biol 2017; 37:823-835. [PMID: 28336561 DOI: 10.1161/atvbaha.117.309253] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 03/08/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) is a collagen receptor that belongs to the inhibitory immunoreceptor tyrosine-based inhibition motif-containing receptor family. It is an inhibitor of signaling via the immunoreceptor tyrosine-based activation motif-containing collagen receptor complex, glycoprotein VI-FcRγ-chain. It is expressed on hematopoietic cells, including immature megakaryocytes, but is not detectable on platelets. Although the inhibitory function of LAIR-1 has been described in leukocytes, its physiological role in megakaryocytes and in particular in platelet formation has not been explored. In this study, we investigate the role of LAIR-1 in megakaryocyte development and platelet production by generating LAIR-1-deficient mice. APPROACH AND RESULTS Mice lacking LAIR-1 exhibit a significant increase in platelet counts, a prolonged platelet half-life in vivo, and increased proplatelet formation in vitro. Interestingly, platelets from LAIR-1-deficient mice exhibit an enhanced reactivity to collagen and the glycoprotein VI-specific agonist collagen-related peptide despite not expressing LAIR-1, and mice showed enhanced thrombus formation in the carotid artery after ferric chloride injury. Targeted deletion of LAIR-1 in mice results in an increase in signaling downstream of the glycoprotein VI-FcRγ-chain and integrin αIIbβ3 in megakaryocytes because of enhanced Src family kinase activity. CONCLUSIONS Findings from this study demonstrate that ablation of LAIR-1 in megakaryocytes leads to increased Src family kinase activity and downstream signaling in response to collagen that is transmitted to platelets, rendering them hyper-reactive specifically to agonists that signal through Syk tyrosine kinases, but not to G-protein-coupled receptors.
Collapse
Affiliation(s)
- Christopher W Smith
- From the Institute of Cardiovascular Sciences, College of Medical and Dental Sciences (C.W.S., S.G.T., Z.R., P.P., M.B., M.L., Y.A.S., S.P.W., A.M.), and Institute of Applied Health Research, College of Medical and Dental Sciences (D.B.), University of Birmingham, United Kingdom; and Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, the Netherlands (L.M.)
| | - Steven G Thomas
- From the Institute of Cardiovascular Sciences, College of Medical and Dental Sciences (C.W.S., S.G.T., Z.R., P.P., M.B., M.L., Y.A.S., S.P.W., A.M.), and Institute of Applied Health Research, College of Medical and Dental Sciences (D.B.), University of Birmingham, United Kingdom; and Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, the Netherlands (L.M.)
| | - Zaher Raslan
- From the Institute of Cardiovascular Sciences, College of Medical and Dental Sciences (C.W.S., S.G.T., Z.R., P.P., M.B., M.L., Y.A.S., S.P.W., A.M.), and Institute of Applied Health Research, College of Medical and Dental Sciences (D.B.), University of Birmingham, United Kingdom; and Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, the Netherlands (L.M.)
| | - Pushpa Patel
- From the Institute of Cardiovascular Sciences, College of Medical and Dental Sciences (C.W.S., S.G.T., Z.R., P.P., M.B., M.L., Y.A.S., S.P.W., A.M.), and Institute of Applied Health Research, College of Medical and Dental Sciences (D.B.), University of Birmingham, United Kingdom; and Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, the Netherlands (L.M.)
| | - Maxwell Byrne
- From the Institute of Cardiovascular Sciences, College of Medical and Dental Sciences (C.W.S., S.G.T., Z.R., P.P., M.B., M.L., Y.A.S., S.P.W., A.M.), and Institute of Applied Health Research, College of Medical and Dental Sciences (D.B.), University of Birmingham, United Kingdom; and Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, the Netherlands (L.M.)
| | - Marie Lordkipanidzé
- From the Institute of Cardiovascular Sciences, College of Medical and Dental Sciences (C.W.S., S.G.T., Z.R., P.P., M.B., M.L., Y.A.S., S.P.W., A.M.), and Institute of Applied Health Research, College of Medical and Dental Sciences (D.B.), University of Birmingham, United Kingdom; and Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, the Netherlands (L.M.)
| | - Danai Bem
- From the Institute of Cardiovascular Sciences, College of Medical and Dental Sciences (C.W.S., S.G.T., Z.R., P.P., M.B., M.L., Y.A.S., S.P.W., A.M.), and Institute of Applied Health Research, College of Medical and Dental Sciences (D.B.), University of Birmingham, United Kingdom; and Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, the Netherlands (L.M.)
| | - Linde Meyaard
- From the Institute of Cardiovascular Sciences, College of Medical and Dental Sciences (C.W.S., S.G.T., Z.R., P.P., M.B., M.L., Y.A.S., S.P.W., A.M.), and Institute of Applied Health Research, College of Medical and Dental Sciences (D.B.), University of Birmingham, United Kingdom; and Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, the Netherlands (L.M.)
| | - Yotis A Senis
- From the Institute of Cardiovascular Sciences, College of Medical and Dental Sciences (C.W.S., S.G.T., Z.R., P.P., M.B., M.L., Y.A.S., S.P.W., A.M.), and Institute of Applied Health Research, College of Medical and Dental Sciences (D.B.), University of Birmingham, United Kingdom; and Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, the Netherlands (L.M.)
| | - Steve P Watson
- From the Institute of Cardiovascular Sciences, College of Medical and Dental Sciences (C.W.S., S.G.T., Z.R., P.P., M.B., M.L., Y.A.S., S.P.W., A.M.), and Institute of Applied Health Research, College of Medical and Dental Sciences (D.B.), University of Birmingham, United Kingdom; and Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, the Netherlands (L.M.)
| | - Alexandra Mazharian
- From the Institute of Cardiovascular Sciences, College of Medical and Dental Sciences (C.W.S., S.G.T., Z.R., P.P., M.B., M.L., Y.A.S., S.P.W., A.M.), and Institute of Applied Health Research, College of Medical and Dental Sciences (D.B.), University of Birmingham, United Kingdom; and Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, the Netherlands (L.M.).
| |
Collapse
|
39
|
Poulter NS, Pollitt AY, Owen DM, Gardiner EE, Andrews RK, Shimizu H, Ishikawa D, Bihan D, Farndale RW, Moroi M, Watson SP, Jung SM. Clustering of glycoprotein VI (GPVI) dimers upon adhesion to collagen as a mechanism to regulate GPVI signaling in platelets. J Thromb Haemost 2017; 15:549-564. [PMID: 28058806 PMCID: PMC5347898 DOI: 10.1111/jth.13613] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Indexed: 01/01/2023]
Abstract
Essentials Dimeric high-affinity collagen receptor glycoprotein VI (GPVI) is present on resting platelets. Spatio-temporal organization of platelet GPVI-dimers was evaluated using advanced microscopy. Upon platelet adhesion to collagenous substrates, GPVI-dimers coalesce to form clusters. Clustering of GPVI-dimers may increase avidity and facilitate platelet activation SUMMARY: Background Platelet glycoprotein VI (GPVI) binding to subendothelial collagen exposed upon blood vessel injury initiates thrombus formation. Dimeric GPVI has high affinity for collagen, and occurs constitutively on resting platelets. Objective To identify higher-order oligomerization (clustering) of pre-existing GPVI dimers upon interaction with collagen as a mechanism to initiate GPVI-mediated signaling. Methods GPVI was located by use of fluorophore-conjugated GPVI dimer-specific Fab (antigen-binding fragment). The tested substrates include Horm collagen I fibers, soluble collagen III, GPVI-specific collagen peptides, and fibrinogen. GPVI dimer clusters on the platelet surface interacting with these substrates were visualized with complementary imaging techniques: total internal reflection fluorescence microscopy to monitor real-time interactions, and direct stochastic optical reconstruction microscopy (dSTORM), providing relative quantification of GPVI cluster size and density. Confocal microscopy was used to locate GPVI dimer clusters, glycoprotein Ib, integrin α2 β1 , and phosphotyrosine. Results Upon platelet adhesion to all collagenous substrates, GPVI dimers coalesced to form clusters; notably clusters formed along the fibers of Horm collagen. dSTORM revealed that GPVI density within clusters depended on the substrate, collagen III being the most effective. Clusters on fibrinogen-adhered platelets were much smaller and more numerous; whether these are pre-existing oligomers of GPVI dimers or fibrinogen-induced is not clear. Some GPVI dimer clusters colocalized with areas of phosphotyrosine, indicative of signaling activity. Integrin α2 β1 was localized to collagen fibers close to GPVI dimer clusters. GPVI clustering depends on a dynamic actin cytoskeleton. Conclusions Platelet adhesion to collagen induces GPVI dimer clustering. GPVI clustering increases both avidity for collagen and the proximity of GPVI-associated signaling molecules, which may be crucial for the initiation and persistence of signaling.
Collapse
Affiliation(s)
- N. S. Poulter
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Centre for Membrane Proteins and Receptors (COMPARE)College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - A. Y. Pollitt
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Present address: Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingRG6 6ASUK
| | - D. M. Owen
- Department of Physics and Randall Division of Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - E. E. Gardiner
- Department of Cancer Biology and TherapeuticsJohn Curtin School of Medical ResearchAustralian National UniversityCanberraACTAustralia
| | - R. K. Andrews
- Australian Centre for Blood DiseasesMonash UniversityMelbourneVictoriaAustralia
| | - H. Shimizu
- Research DepartmentChemo‐Sero‐Therapeutic Research InstituteKaketsukenKumamotoJapan
| | - D. Ishikawa
- Research DepartmentChemo‐Sero‐Therapeutic Research InstituteKaketsukenKumamotoJapan
| | - D. Bihan
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - R. W. Farndale
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - M. Moroi
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - S. P. Watson
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Centre for Membrane Proteins and Receptors (COMPARE)College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - S. M. Jung
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| |
Collapse
|
40
|
Technologies for Proteome-Wide Discovery of Extracellular Host-Pathogen Interactions. J Immunol Res 2017; 2017:2197615. [PMID: 28321417 PMCID: PMC5340944 DOI: 10.1155/2017/2197615] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 01/19/2017] [Indexed: 12/26/2022] Open
Abstract
Pathogens have evolved unique mechanisms to breach the cell surface barrier and manipulate the host immune response to establish a productive infection. Proteins exposed to the extracellular environment, both cell surface-expressed receptors and secreted proteins, are essential targets for initial invasion and play key roles in pathogen recognition and subsequent immunoregulatory processes. The identification of the host and pathogen extracellular molecules and their interaction networks is fundamental to understanding tissue tropism and pathogenesis and to inform the development of therapeutic strategies. Nevertheless, the characterization of the proteins that function in the host-pathogen interface has been challenging, largely due to the technical challenges associated with detection of extracellular protein interactions. This review discusses available technologies for the high throughput study of extracellular protein interactions between pathogens and their hosts, with a focus on mammalian viruses and bacteria. Emerging work illustrates a rich landscape for extracellular host-pathogen interaction and points towards the evolution of multifunctional pathogen-encoded proteins. Further development and application of technologies for genome-wide identification of extracellular protein interactions will be important in deciphering functional host-pathogen interaction networks, laying the foundation for development of novel therapeutics.
Collapse
|
41
|
Abstract
In this issue of Blood, Zhou et al reported the high-resolution structure of the collagen-activated osteoclast-associated receptor (OSCAR) bound to a collagen model peptide. Together with binding studies, the results confirm a novel recognition mechanism for collagen by immunoglobulin-like motifs.
Collapse
|
42
|
Collagen interactions: Drug design and delivery. Adv Drug Deliv Rev 2016; 97:69-84. [PMID: 26631222 DOI: 10.1016/j.addr.2015.11.013] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/19/2015] [Accepted: 11/20/2015] [Indexed: 12/25/2022]
Abstract
Collagen is a major component in a wide range of drug delivery systems and biomaterial applications. Its basic physical and structural properties, together with its low immunogenicity and natural turnover, are keys to its biocompatibility and effectiveness. In addition to its material properties, the collagen triple-helix interacts with a large number of molecules that trigger biological events. Collagen interactions with cell surface receptors regulate many cellular processes, while interactions with other ECM components are critical for matrix structure and remodeling. Collagen also interacts with enzymes involved in its biosynthesis and degradation, including matrix metalloproteinases. Over the past decade, much information has been gained about the nature and specificity of collagen interactions with its partners. These studies have defined collagen sequences responsible for binding and the high-resolution structures of triple-helical peptides bound to its natural binding partners. Strategies to target collagen interactions are already being developed, including the use of monoclonal antibodies to interfere with collagen fibril formation and the use of triple-helical peptides to direct liposomes to melanoma cells. The molecular information about collagen interactions will further serve as a foundation for computational studies to design small molecules that can interfere with specific interactions or target tumor cells. Intelligent control of collagen biological interactions within a material context will expand the effectiveness of collagen-based drug delivery.
Collapse
|
43
|
Structural basis of collagen recognition by human osteoclast-associated receptor and design of osteoclastogenesis inhibitors. Proc Natl Acad Sci U S A 2016; 113:1038-43. [PMID: 26744311 DOI: 10.1073/pnas.1522572113] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Human osteoclast-associated receptor (OSCAR) is an immunoglobulin (Ig)-like collagen receptor that is up-regulated on osteoclasts during osteoclastogenesis and is expressed in a range of myeloid cells. As a member of the leukocyte receptor complex family of proteins, OSCAR shares a high degree of sequence and structural homology with other collagen receptors of this family, including glycoprotein VI, leukocyte-associated Ig-like receptor-1, and leukocyte Ig-like receptor B4, but recognizes a unique collagen sequence. Here, we present the crystal structures of OSCAR in its free form and in complex with a triple-helical collagen-like peptide (CLP). These structures reveal that the CLP peptide binds only one of the two Ig-like domains, the membrane-proximal domain (domain 2) of OSCAR, with the middle and trailing chain burying a total of 661 Å(2) of solvent-accessible collagen surface. This binding mode is facilitated by the unusual topography of the OSCAR protein, which displays an obtuse interdomain angle and a rotation of domain 2 relative to the membrane-distal domain 1. Moreover, the binding of the CLP to OSCAR appears to be mediated largely by tyrosine residues and conformational changes at a shallow Phe pocket. Furthermore, we investigated CLP peptides as inhibitors of osteoclastogenesis and found that a peptide length of 40 amino acids is required to ensure adequate inhibition of osteoclastogenesis in vitro. These findings provide valuable structural insights into the mode of collagen recognition by OSCAR and into the use of synthetic peptide matrikines for osteoclastogenesis inhibition.
Collapse
|
44
|
Karo-Atar D, Bordowitz A, Wand O, Pasmanik-Chor M, Fernandez IE, Itan M, Frenkel R, Herbert DR, Finkelman FD, Eickelberg O, Munitz A. A protective role for IL-13 receptor α 1 in bleomycin-induced pulmonary injury and repair. Mucosal Immunol 2016; 9:240-53. [PMID: 26153764 PMCID: PMC4703942 DOI: 10.1038/mi.2015.56] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 05/14/2015] [Indexed: 02/04/2023]
Abstract
Molecular mechanisms that regulate lung repair vs. progressive scarring in pulmonary fibrosis remain elusive. Interleukin (IL)-4 and IL-13 are pro-fibrotic cytokines that share common receptor chains including IL-13 receptor (R) α1 and are key pharmacological targets in fibrotic diseases. However, the roles of IL-13Rα1 in mediating lung injury/repair are unclear. We report dysregulated levels of IL-13 receptors in the lungs of bleomycin-treated mice and to some extent in idiopathic pulmonary fibrosis patients. Transcriptional profiling demonstrated an epithelial cell-associated gene signature that was homeostatically dependent on IL-13Rα1 expression. IL-13Rα1 regulated a striking array of genes in the lung following bleomycin administration and Il13ra1 deficiency resulted in exacerbated bleomycin-induced disease. Increased pathology in bleomycin-treated Il13ra1(-/-) mice was due to IL-13Rα1 expression in structural and hematopoietic cells but not due to increased responsiveness to IL-17, IL-4, IL-13, increased IL-13Rα2 or type 1 IL-4R signaling. These data highlight underappreciated protective roles for IL-13Rα1 in lung injury and homeostasis.
Collapse
Affiliation(s)
- D Karo-Atar
- Department of Clinical Microbiology and Immunology, The Sackler School of Medicine, The Tel-Aviv University, Ramat Aviv, Israel
| | - A Bordowitz
- Department of Clinical Microbiology and Immunology, The Sackler School of Medicine, The Tel-Aviv University, Ramat Aviv, Israel
| | - O Wand
- Department of Clinical Microbiology and Immunology, The Sackler School of Medicine, The Tel-Aviv University, Ramat Aviv, Israel
| | - M Pasmanik-Chor
- Bioinformatics Unit, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - I E Fernandez
- Comprehensive Pneumology Center, Ludwig Maximilians University, University Hospital Grosshadern, and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - M Itan
- Department of Clinical Microbiology and Immunology, The Sackler School of Medicine, The Tel-Aviv University, Ramat Aviv, Israel
| | - R Frenkel
- Department of Math, Physics and Computer Science, University of Cincinnati, Cincinnati, Ohio, USA
| | - D R Herbert
- Division of Experimental Medicine, University of California, San Francisco, California, USA
| | - F D Finkelman
- Division of Allergy, Immunology and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA,Department of Medicine, Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - O Eickelberg
- Comprehensive Pneumology Center, Ludwig Maximilians University, University Hospital Grosshadern, and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - A Munitz
- Department of Clinical Microbiology and Immunology, The Sackler School of Medicine, The Tel-Aviv University, Ramat Aviv, Israel,()
| |
Collapse
|
45
|
A LAIR1 insertion generates broadly reactive antibodies against malaria variant antigens. Nature 2015; 529:105-109. [PMID: 26700814 PMCID: PMC4869849 DOI: 10.1038/nature16450] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/16/2015] [Indexed: 01/04/2023]
Abstract
Plasmodium falciparum antigens expressed on the surface of infected erythrocytes are important targets of naturally acquired immunity against malaria, but their high number and variability provide the pathogen with a powerful means of escape from host antibodies1–4. Although broadly reactive antibodies against these antigens could be useful as therapeutics and in vaccine design, their identification has proven elusive. Here, we report the isolation of human monoclonal antibodies that recognize erythrocytes infected by different P. falciparum isolates and opsonize these cells by binding to members of the RIFIN family. These antibodies acquired broad reactivity through a novel mechanism of insertion of a large DNA fragment between the V and DJ segments. The insert, which is both necessary and sufficient for binding to RIFINs, encodes the entire 100 amino acid collagen-binding domain of LAIR-1, an Ig superfamily inhibitory receptor encoded on chromosome 19. In each of the two donors studied, the antibodies are produced by a single expanded B cell clone and carry distinct somatic mutations in the LAIR-1 domain that abolish binding to collagen and increase binding to infected erythrocytes. These findings illustrate, with a biologically relevant example, a novel mechanism of antibody diversification by interchromosomal DNA transposition and demonstrate the existence of conserved epitopes that may be suitable candidates for the development of a malaria vaccine.
Collapse
|
46
|
Structural basis for collagen recognition by the immune receptor OSCAR. Blood 2015; 127:529-37. [PMID: 26552697 DOI: 10.1182/blood-2015-08-667055] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/02/2015] [Indexed: 12/18/2022] Open
Abstract
The osteoclast-associated receptor (OSCAR) is a collagen-binding immune receptor with important roles in dendritic cell maturation and activation of inflammatory monocytes as well as in osteoclastogenesis. The crystal structure of the OSCAR ectodomain is presented, both free and in complex with a consensus triple-helical peptide (THP). The structures revealed a collagen-binding site in each immunoglobulin-like domain (D1 and D2). The THP binds near a predicted collagen-binding groove in D1, but a more extensive interaction with D2 is facilitated by the unusually wide D1-D2 interdomain angle in OSCAR. Direct binding assays, combined with site-directed mutagenesis, confirm that the primary collagen-binding site in OSCAR resides in D2, in marked contrast to the related collagen receptors, glycoprotein VI (GPVI) and leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1). Monomeric OSCAR D1D2 binds to the consensus THP with a KD of 28 µM measured in solution, but shows a higher affinity (KD 1.5 μM) when binding to a solid-phase THP, most likely due to an avidity effect. These data suggest a 2-stage model for the interaction of OSCAR with a collagen fibril, with transient, low-affinity interactions initiated by the membrane-distal D1, followed by firm adhesion to the primary binding site in D2.
Collapse
|
47
|
Ero-Tolliver IA, Hudson BG, Bhave G. The Ancient Immunoglobulin Domains of Peroxidasin Are Required to Form Sulfilimine Cross-links in Collagen IV. J Biol Chem 2015; 290:21741-8. [PMID: 26178375 PMCID: PMC4571896 DOI: 10.1074/jbc.m115.673996] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 07/13/2015] [Indexed: 02/02/2023] Open
Abstract
The collagen IV sulfilimine cross-link and its catalyzing enzyme, peroxidasin, represent a dyad critical for tissue development, which is conserved throughout the animal kingdom. Peroxidasin forms novel sulfilimine bonds between opposing methionine and hydroxylysine residues to structurally reinforce the collagen IV scaffold, a function critical for basement membrane and tissue integrity. However, the molecular mechanism underlying cross-link formation remains unclear. In this work, we demonstrate that the catalytic domain of peroxidasin and its immunoglobulin (Ig) domains are required for efficient sulfilimine bond formation. Thus, these molecular features underlie the evolutionarily conserved function of peroxidasin in tissue development and integrity and distinguish peroxidasin from other peroxidases, such as myeloperoxidase (MPO) and eosinophil peroxidase (EPO).
Collapse
Affiliation(s)
- Isi A Ero-Tolliver
- From the Division of Nephrology and Hypertension and Department of Medicine, Center for Matrix Biology
| | - Billy G Hudson
- From the Division of Nephrology and Hypertension and Department of Medicine, Center for Matrix Biology, Departments of Biochemistry and Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Gautam Bhave
- From the Division of Nephrology and Hypertension and Department of Medicine, Center for Matrix Biology,
| |
Collapse
|
48
|
Sun S, Jiao Y, Wei W, Postlethwaite AE, Gu W, Sun D. Comparison of LAIR-1 genetic pathways in murine vs human internal organs. Gene 2014; 552:140-5. [PMID: 25234728 DOI: 10.1016/j.gene.2014.09.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 08/20/2014] [Accepted: 09/14/2014] [Indexed: 01/08/2023]
Abstract
Growing evidence suggests that defective expression or dysfunction of LAIR-1, a novel immunoinhibitory receptor for collagen, is closely associated with some autoimmune diseases, cancers, as well as viral infections. We analyzed the variation of LAIR-1 genetic pathways in murine versus human internal organs, including the lung and brain. The results showed that, under physiological conditions, LAIR-1 links more closely to the common genes in mouse than in human, which poses tissue specificity. It means that mice experimental data in relation to the role of LAIR-1 immune regulation may be overestimated when applied to assess human conditions. Moreover, we found that the in vivo interaction of LAIR-1 with LAIR-2 rarely occurs, implying that the species difference in LAIR-1 genetic pathways could not be primarily attributed to the existence of human LAIR-2. In summary, this study opens the door for insight into LAIR-1 functions inside the human body, and raises concern as to extrapolative credibility of the murine model in biomedical research.
Collapse
Affiliation(s)
- Shuqiu Sun
- National Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China; Department of Orthopedic Surgery and BME, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yan Jiao
- Department of Orthopedic Surgery and BME, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Wei Wei
- National Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China
| | - Arnold E Postlethwaite
- Division of Connective Tissue Diseases, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Veterans Affairs Medical Center, Memphis TN 38104, USA
| | - Weikuan Gu
- Department of Orthopedic Surgery and BME, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Veterans Affairs Medical Center, Memphis TN 38104, USA.
| | - Dianjun Sun
- National Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
49
|
Sangaletti S, Tripodo C, Vitali C, Portararo P, Guarnotta C, Casalini P, Cappetti B, Miotti S, Pinciroli P, Fuligni F, Fais F, Piccaluga PP, Colombo MP. Defective stromal remodeling and neutrophil extracellular traps in lymphoid tissues favor the transition from autoimmunity to lymphoma. Cancer Discov 2014; 4:110-29. [PMID: 24189145 DOI: 10.1158/2159-8290.cd-13-0276] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Altered expression of matricellular proteins can become pathogenic in the presence of persistent perturbations in tissue homeostasis. Here, we show that autoimmunity associated with Fas mutation was exacerbated and transitioned to lymphomagenesis in the absence of SPARC (secreted protein acidic rich in cysteine). The absence of SPARC resulted in defective collagen assembly, with uneven compartmentalization of lymphoid and myeloid populations within secondary lymphoid organs (SLO), and faulty delivery of inhibitory signals from the extracellular matrix. These conditions promoted aberrant interactions between neutrophil extracellular traps and CD5(+) B cells, which underwent malignant transformation due to defective apoptosis under the pressure of neutrophil-derived trophic factors and NF-κB activation. Furthermore, this model of defective stromal remodeling during lymphomagenesis correlates with human lymphomas arising in a SPARC-defective environment, which is prototypical of CD5(+) B-cell chronic lymphocytic leukemia (CLL).
Collapse
Affiliation(s)
- Sabina Sangaletti
- 1Molecular Immunology Unit, 2Molecular Targeting Unit, and 3Molecular Therapies Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan; 4Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo; 5Hematopathology Section, Department of Hematology and Oncology L. and A. Seràgnoli, S. Orsola-Malpighi Hospital, University of Bologna, Bologna; and 6Human Anatomy Section, Department of Experimental Medicine, University of Genova, Genova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Zhang Y, Lv K, Zhang C, Jin B, Zhuang R, Ding Y. The role of LAIR-1 (CD305) in T cells and monocytes/macrophages in patients with rheumatoid arthritis. Cell Immunol 2014; 287:46-52. [DOI: 10.1016/j.cellimm.2013.12.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 11/26/2013] [Accepted: 12/05/2013] [Indexed: 01/21/2023]
|