151
|
Cerny O, Godlee C, Tocci R, Cross NE, Shi H, Williamson JC, Alix E, Lehner PJ, Holden DW. CD97 stabilises the immunological synapse between dendritic cells and T cells and is targeted for degradation by the Salmonella effector SteD. PLoS Pathog 2021; 17:e1009771. [PMID: 34314469 PMCID: PMC8345877 DOI: 10.1371/journal.ppat.1009771] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/06/2021] [Accepted: 06/29/2021] [Indexed: 11/19/2022] Open
Abstract
The Salmonella enterica effector SteD depletes mature MHC class II (mMHCII) molecules from the surface of infected antigen-presenting cells through ubiquitination of the cytoplasmic tail of the mMHCII β chain. This requires the Nedd4 family HECT E3 ubiquitin ligase Wwp2 and a tumor-suppressing transmembrane protein adaptor Tmem127. Here, through a proteomic screen of dendritic cells, we found that SteD targets the plasma membrane protein CD97 for degradation by a similar mechanism. SteD enhanced ubiquitination of CD97 on K555 and mutation of this residue eliminated the effect of SteD on CD97 surface levels. We showed that CD97 localises to and stabilises the immunological synapse between dendritic cells and T cells. Removal of CD97 by SteD inhibited dendritic cell-T cell interactions and reduced T cell activation, independently of its effect on MHCII. Therefore, SteD suppresses T cell immunity by two distinct processes.
Collapse
Affiliation(s)
- Ondrej Cerny
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
- * E-mail: (OC); (DWH)
| | - Camilla Godlee
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Romina Tocci
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Nancy E. Cross
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Haoran Shi
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - James C. Williamson
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Cambridge, United Kingdom
| | - Eric Alix
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Paul J. Lehner
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Cambridge, United Kingdom
| | - David W. Holden
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
- * E-mail: (OC); (DWH)
| |
Collapse
|
152
|
Bonilha CS, Benson RA, Scales HE, Brewer JM, Garside P. Junctional adhesion molecule-A on dendritic cells regulates Th1 differentiation. Immunol Lett 2021; 235:32-40. [PMID: 34000305 DOI: 10.1016/j.imlet.2021.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/26/2021] [Accepted: 05/08/2021] [Indexed: 02/06/2023]
Abstract
The junctional adhesion molecule-A (JAM-A) is an adhesion molecule present in the surface of several cell types, such as endothelial cells and leukocytes as well as Dendritic Cells (DC). Given the potential relevance of JAM-A in diverse pathological conditions such as inflammatory diseases and cancer, we investigated the role of JAM-A in CD4+ T cell priming. We demonstrate that JAM-A is present in the immunological synapse formed between T cells and DC during priming. Furthermore, an antagonistic anti-JAM-A mAb could disrupt the interaction between CD4+ T cell and DC. Antagonism of JAM-A also attenuated T cell activation and proliferation with a decrease in T-bet expression and increased IL-6 and IL-17 secretion. These findings demonstrate a functional role for JAM-A in interactions between CD4+ T cells and DCs during T cell priming as a positive regulator of Th1 differentiation.
Collapse
Affiliation(s)
- Caio S Bonilha
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK.
| | - Robert A Benson
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Hannah E Scales
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - James M Brewer
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Paul Garside
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK.
| |
Collapse
|
153
|
Berger RML, Weck JM, Kempe SM, Hill O, Liedl T, Rädler JO, Monzel C, Heuer-Jungemann A. Nanoscale FasL Organization on DNA Origami to Decipher Apoptosis Signal Activation in Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101678. [PMID: 34057291 DOI: 10.1002/smll.202101678] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/13/2021] [Indexed: 05/27/2023]
Abstract
Cell signaling is initiated by characteristic protein patterns in the plasma membrane, but tools to decipher their molecular organization and activation are hitherto lacking. Among the well-known signaling pattern is the death inducing signaling complex with a predicted hexagonal receptor architecture. To probe this architecture, DNA origami-based nanoagents with nanometer precise arrangements of the death receptor ligand FasL are introduced and presented to cells. Mimicking different receptor geometries, these nanoagents act as signaling platforms inducing fastest time-to-death kinetics for hexagonal FasL arrangements with 10 nm inter-molecular spacing. Compared to naturally occurring soluble FasL, this trigger is faster and 100× more efficient. Nanoagents with different spacing, lower FasL number or higher coupling flexibility impede signaling. The results present DNA origami as versatile signaling scaffolds exhibiting unprecedented control over molecular number and geometry. They define molecular benchmarks in apoptosis signal initiation and constitute a new strategy to drive particular cell responses.
Collapse
Affiliation(s)
- Ricarda M L Berger
- Faculty of Physics and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
| | - Johann M Weck
- Max Planck Institute of Biochemistry and Center for Nanoscience (CeNS), Ludwig-Maximilians-University, Am Klopferspitz 18, 82152, Martinsried, Germany
| | - Simon M Kempe
- Faculty of Physics and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
| | - Oliver Hill
- Apogenix AG, University of Heidelberg, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Tim Liedl
- Faculty of Physics and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
| | - Joachim O Rädler
- Faculty of Physics and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
| | - Cornelia Monzel
- Experimental Medical Physics, Heinrich-Heine University, Universitätsstrasse 1, 40225, Düsseldorf, Germany
| | - Amelie Heuer-Jungemann
- Max Planck Institute of Biochemistry and Center for Nanoscience (CeNS), Ludwig-Maximilians-University, Am Klopferspitz 18, 82152, Martinsried, Germany
| |
Collapse
|
154
|
Bezverbnaya K, Moogk D, Cummings D, Baker CL, Aarts C, Denisova G, Sun M, McNicol JD, Turner RC, Rullo AF, Foley SR, Bramson JL. Development of a B-cell maturation antigen-specific T-cell antigen coupler receptor for multiple myeloma. Cytotherapy 2021; 23:820-832. [PMID: 34217618 DOI: 10.1016/j.jcyt.2021.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS T cells engineered with synthetic receptors have delivered powerful therapeutic results for patients with relapsed/refractory hematologic malignancies. The authors have recently described the T-cell antigen coupler (TAC) receptor, which co-opts the endogenous T-cell receptor (TCR) and activates engineered T cells in an HLA-independent manner. Here the authors describe the evolution of a next-generation TAC receptor with a focus on developing a TAC-engineered T cell for multiple myeloma. METHODS To optimize the TAC scaffold, the authors employed a bona fide antigen-binding domain derived from the B-cell maturation antigen-specific monoclonal antibody C11D5.3, which has been used successfully in the clinic. The authors first tested humanized versions of the UCHT1 domain, which is used by the TAC to co-opt the TCR. The authors further discovered that the signal peptide affected surface expression of the TAC receptor. Higher density of the TAC receptor enhanced target binding in vitro, which translated into higher levels of Lck at the immunological synapse and stronger proliferation when only receptor-ligand interactions were present. RESULTS The authors observed that the humanized UCHT1 improved surface expression and in vivo efficacy. Using TAC T cells derived from both healthy donors and multiple myeloma patients, the authors determined that despite the influence of receptor density on early activation events and effector function, receptor density did not impact late effector functions in vitro, nor did the receptor density affect in vivo efficacy. CONCLUSIONS The modifications to the TAC scaffold described herein represent an important step in the evolution of this technology, which tolerates a range of expression levels without impacting therapeutic efficacy.
Collapse
Affiliation(s)
- Ksenia Bezverbnaya
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Duane Moogk
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Derek Cummings
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Christopher L Baker
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Craig Aarts
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Galina Denisova
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Michael Sun
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Jamie D McNicol
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Rebecca C Turner
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Anthony F Rullo
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - S Ronan Foley
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada; Juravinski Hospital and Cancer Centre, Hamilton, Canada
| | - Jonathan L Bramson
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada.
| |
Collapse
|
155
|
GABAergic signaling by cells of the immune system: more the rule than the exception. Cell Mol Life Sci 2021; 78:5667-5679. [PMID: 34152447 PMCID: PMC8316187 DOI: 10.1007/s00018-021-03881-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/17/2021] [Accepted: 06/11/2021] [Indexed: 11/23/2022]
Abstract
Gamma-aminobutyric acid (GABA) is best known as an essential neurotransmitter in the evolved central nervous system (CNS) of vertebrates. However, GABA antedates the development of the CNS as a bioactive molecule in metabolism and stress-coupled responses of prokaryotes, invertebrates and plants. Here, we focus on the emerging findings of GABA signaling in the mammalian immune system. Recent reports show that mononuclear phagocytes and lymphocytes, for instance dendritic cells, microglia, T cells and NK cells, express a GABAergic signaling machinery. Mounting evidence shows that GABA receptor signaling impacts central immune functions, such as cell migration, cytokine secretion, immune cell activation and cytotoxic responses. Furthermore, the GABAergic signaling machinery of leukocytes is implicated in responses to microbial infection and is co-opted by protozoan parasites for colonization of the host. Peripheral GABA signaling is also implicated in inflammatory conditions and diseases, such as type 1 diabetes, rheumatoid arthritis and cancer cell metastasis. Adding to its role in neurotransmission, growing evidence shows that the non-proteinogenic amino acid GABA acts as an intercellular signaling molecule in the immune system and, as an interspecies signaling molecule in host–microbe interactions. Altogether, the data raise the assumption of conserved GABA signaling in a broad range of mammalian cells and diversification of function in the immune system.
Collapse
|
156
|
Felce SL, Farnie G, Dustin ML, Felce JH. RNA-Seq analysis of early transcriptional responses to activation in the leukaemic Jurkat E6.1 T cell line. Wellcome Open Res 2021; 5:42. [PMID: 36865034 PMCID: PMC9971649 DOI: 10.12688/wellcomeopenres.15748.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2021] [Indexed: 11/20/2022] Open
Abstract
Background: The leukaemia-derived Jurkat E6.1 cell line has been used as a model T cell in the study of many aspects of T cell biology, most notably activation in response to T cell receptor (TCR) engagement. Methods: We present whole-transcriptome RNA-Sequencing data for Jurkat E6.1 cells in the resting state and two hours post-activation via TCR and CD28. We compare early transcriptional responses in the presence and absence of the chemokines CXCL12 and CCL19, and perform a basic comparison between observed transcriptional responses in Jurkat E6.1 cells and those in primary human T cells using publicly deposited data. Results: Jurkat E6.1 cells have many of the hallmarks of standard T cell transcriptional responses to activation, but lack most of the depth of responses in primary cells. Conclusions: These data indicate that Jurkat E6.1 cells hence represent only a highly simplified model of early T cell transcriptional responses.
Collapse
Affiliation(s)
- Suet Ling Felce
- Structural Genomics Consortium, Botnar Research Centre, NDORMS, University of Oxford, Oxford, OX3 7LD, UK
| | - Gillian Farnie
- Structural Genomics Consortium, Botnar Research Centre, NDORMS, University of Oxford, Oxford, OX3 7LD, UK
| | - Michael L. Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - James H. Felce
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| |
Collapse
|
157
|
Zhang Y, Liu Q, Yang S, Liao Q. CD58 Immunobiology at a Glance. Front Immunol 2021; 12:705260. [PMID: 34168659 PMCID: PMC8218816 DOI: 10.3389/fimmu.2021.705260] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 05/24/2021] [Indexed: 01/12/2023] Open
Abstract
The glycoprotein CD58, also known as lymphocyte-function antigen 3 (LFA-3), is a costimulatory receptor distributed on a broad range of human tissue cells. Its natural ligand CD2 is primarily expressed on the surface of T/NK cells. The CD2-CD58 interaction is an important component of the immunological synapse (IS) that induces activation and proliferation of T/NK cells and triggers a series of intracellular signaling in T/NK cells and target cells, respectively, in addition to promoting cell adhesion and recognition. Furthermore, a soluble form of CD58 (sCD58) is also present in cellular supernatant in vitro and in local tissues in vivo. The sCD58 is involved in T/NK cell-mediated immune responses as an immunosuppressive factor by affecting CD2-CD58 interaction. Altered accumulation of sCD58 may lead to immunosuppression of T/NK cells in the tumor microenvironment, allowing sCD58 as a novel immunotherapeutic target. Recently, the crucial roles of costimulatory molecule CD58 in immunomodulation seem to be reattracting the interests of investigators. In particular, the CD2-CD58 interaction is involved in the regulation of antiviral responses, inflammatory responses in autoimmune diseases, immune rejection of transplantation, and immune evasion of tumor cells. In this review, we provide a comprehensive summary of CD58 immunobiology.
Collapse
Affiliation(s)
- Yalu Zhang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Qiaofei Liu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Sen Yang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Quan Liao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
158
|
Novosad J, Krčmová I. Evolution of our view on the IgE molecule role in bronchial asthma and the clinical effect of its modulation by omalizumab: Where do we stand today? Int J Immunopathol Pharmacol 2021; 34:2058738420942386. [PMID: 32689848 PMCID: PMC7375718 DOI: 10.1177/2058738420942386] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bronchial asthma is a heterogeneous disease whose definition and treatment are based on evidence of variable airway obstruction and airway inflammation. Despite the enormous increase in the amount of information on the pathogenesis of this disease, diagnosis is still an unresolved problem, as we still lack sensitive and specific biomarkers. On the other hand, at the turn of the 20th and 21st century, there was a rapid development of therapeutic modalities based on the principle of biological therapy. The first authorized drug matching these characteristics was omalizumab – a monoclonal antibody directed against immunoglobulin E (IgE). It has been used for the treatment of severe forms of bronchial asthma for more than 15 years, which is a sufficient time to acquire ways of its effective use and to assess whether the treatment with omalizumab has met our expectations. However, we continue to discover new and surprising facts about the effects of omalizumab treatment which leads to widening of therapeutic indications. In this work, a basic overview of the very complex role of the IgE molecule in the organism (with a special emphasis on allergic asthma) is discussed, and the most important practical and clinical consequences resulting from its modulation by targeted therapy with omalizumab are summarized.
Collapse
Affiliation(s)
- Jakub Novosad
- Institute of Clinical Immunology and Allergology, University Hospital Hradec Králové, Hradec Králové, Czech Republic.,Faculty of Medicine in Hradec Králové, Charles University in Prague, Prague, Czech Republic
| | - Irena Krčmová
- Institute of Clinical Immunology and Allergology, University Hospital Hradec Králové, Hradec Králové, Czech Republic.,Faculty of Medicine in Hradec Králové, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
159
|
Having an Old Friend for Dinner: The Interplay between Apoptotic Cells and Efferocytes. Cells 2021; 10:cells10051265. [PMID: 34065321 PMCID: PMC8161178 DOI: 10.3390/cells10051265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 02/02/2023] Open
Abstract
Apoptosis, the programmed and intentional death of senescent, damaged, or otherwise superfluous cells, is the natural end-point for most cells within multicellular organisms. Apoptotic cells are not inherently damaging, but if left unattended, they can lyse through secondary necrosis. The resulting release of intracellular contents drives inflammation in the surrounding tissue and can lead to autoimmunity. These negative consequences of secondary necrosis are avoided by efferocytosis—the phagocytic clearance of apoptotic cells. Efferocytosis is a product of both apoptotic cells and efferocyte mechanisms, which cooperate to ensure the rapid and complete removal of apoptotic cells. Herein, we review the processes used by apoptotic cells to ensure their timely removal, and the receptors, signaling, and cellular processes used by efferocytes for efferocytosis, with a focus on the receptors and signaling driving this process.
Collapse
|
160
|
CD11c regulates hematopoietic stem and progenitor cells under stress. Blood Adv 2021; 4:6086-6097. [PMID: 33351105 DOI: 10.1182/bloodadvances.2020002504] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/09/2020] [Indexed: 12/28/2022] Open
Abstract
β2 integrins are well-known leukocyte adhesion molecules consisting of 4 members: CD11a-d. Their known biological functions range widely from leukocyte recruitment, phagocytosis, to immunological synapse formation, but the studies have been primarily focused on CD11a and CD11b. CD11c is 1 of the 4 members and is extremely homologous to CD11b. It has been well known as a dendritic cell marker, but the characterization of its function has been limited. We found that CD11c was expressed on the short-term hematopoietic stem cells and multipotent progenitor cells. The lack of CD11c did not affect the number of hematopoietic stem and progenitor cells (HSPCs) in healthy CD11c knockout mice. Different from other β2 integrin members, however, CD11c deficiency was associated with increased apoptosis and significant loss of HSPCs in sepsis and bone marrow transplantation. Although integrins are generally known for their overlapping and redundant roles, we showed that CD11c had a distinct role of regulating the expansion of HSPCs under stress. This study shows that CD11c, a well-known dendritic cell marker, is expressed on HSPCs and serves as their functional regulator. CD11c deficiency leads to the loss of HSPCs via apoptosis in sepsis and bone marrow transplantation.
Collapse
|
161
|
PD-1 suppresses TCR-CD8 cooperativity during T-cell antigen recognition. Nat Commun 2021; 12:2746. [PMID: 33980853 PMCID: PMC8115078 DOI: 10.1038/s41467-021-22965-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 04/09/2021] [Indexed: 12/31/2022] Open
Abstract
Despite the clinical success of blocking its interactions, how PD-1 inhibits T-cell activation is incompletely understood, as exemplified by its potency far exceeding what might be predicted from its affinity for PD-1 ligand-1 (PD-L1). This may be partially attributed to PD-1's targeting the proximal signaling of the T-cell receptor (TCR) and co-stimulatory receptor CD28 via activating Src homology region 2 domain-containing phosphatases (SHPs). Here, we report PD-1 signaling regulates the initial TCR antigen recognition manifested in a smaller spreading area, fewer molecular bonds formed, and shorter bond lifetime of T cell interaction with peptide-major histocompatibility complex (pMHC) in the presence than absence of PD-L1 in a manner dependent on SHPs and Leukocyte C-terminal Src kinase. Our results identify a PD-1 inhibitory mechanism that disrupts the cooperative TCR-pMHC-CD8 trimolecular interaction, which prevents CD8 from augmenting antigen recognition, explaining PD-1's potent inhibitory function and its value as a target for clinical intervention.
Collapse
|
162
|
Tian Z, Liu M, Zhang Y, Wang X. Bispecific T cell engagers: an emerging therapy for management of hematologic malignancies. J Hematol Oncol 2021; 14:75. [PMID: 33941237 PMCID: PMC8091790 DOI: 10.1186/s13045-021-01084-4] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
Harnessing the power of immune cells, especially T cells, to enhance anti-tumor activities has become a promising strategy in clinical management of hematologic malignancies. The emerging bispecific antibodies (BsAbs), which recruit T cells to tumor cells, exemplified by bispecific T cell engagers (BiTEs), have facilitated the development of tumor immunotherapy. Here we discussed the advances and challenges in BiTE therapy developed for the treatment of hematologic malignancies. Blinatumomab, the first BiTE approved for the treatment of acute lymphocytic leukemia (ALL), is appreciated for its high efficacy and safety. Recent studies have focused on improving the efficacy of BiTEs by optimizing treatment regimens and refining the molecular structures of BiTEs. A considerable number of bispecific T cell-recruiting antibodies which are potentially effective in hematologic malignancies have been derived from BiTEs. The elucidation of mechanisms of BiTE action and neonatal techniques used for the construction of BsAbs can improve the treatment of hematological malignancies. This review summarized the features of bispecific T cell-recruiting antibodies for the treatment of hematologic malignancies with special focus on preclinical experiments and clinical studies.
Collapse
Affiliation(s)
- Zheng Tian
- School of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Ming Liu
- Department of Hematology, Shandong Provincial Hospital Affiliated To Shandong University, Shandong First Medical University, No.324, Jingwu Road, Jinan, 250021, Shandong, China.,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China.,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China
| | - Ya Zhang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated To Shandong University, Shandong First Medical University, No.324, Jingwu Road, Jinan, 250021, Shandong, China. .,School of Medicine, Shandong University, Jinan, 250012, Shandong, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China. .,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China. .,National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated To Shandong University, Shandong First Medical University, No.324, Jingwu Road, Jinan, 250021, Shandong, China. .,School of Medicine, Shandong University, Jinan, 250012, Shandong, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China. .,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China. .,National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
163
|
Sanders DW, Jumper CC, Ackerman PJ, Bracha D, Donlic A, Kim H, Kenney D, Castello-Serrano I, Suzuki S, Tamura T, Tavares AH, Saeed M, Holehouse AS, Ploss A, Levental I, Douam F, Padera RF, Levy BD, Brangwynne CP. SARS-CoV-2 requires cholesterol for viral entry and pathological syncytia formation. eLife 2021; 10:e65962. [PMID: 33890572 PMCID: PMC8104966 DOI: 10.7554/elife.65962] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/01/2021] [Indexed: 12/27/2022] Open
Abstract
Many enveloped viruses induce multinucleated cells (syncytia), reflective of membrane fusion events caused by the same machinery that underlies viral entry. These syncytia are thought to facilitate replication and evasion of the host immune response. Here, we report that co-culture of human cells expressing the receptor ACE2 with cells expressing SARS-CoV-2 spike, results in synapse-like intercellular contacts that initiate cell-cell fusion, producing syncytia resembling those we identify in lungs of COVID-19 patients. To assess the mechanism of spike/ACE2-driven membrane fusion, we developed a microscopy-based, cell-cell fusion assay to screen ~6000 drugs and >30 spike variants. Together with quantitative cell biology approaches, the screen reveals an essential role for biophysical aspects of the membrane, particularly cholesterol-rich regions, in spike-mediated fusion, which extends to replication-competent SARS-CoV-2 isolates. Our findings potentially provide a molecular basis for positive outcomes reported in COVID-19 patients taking statins and suggest new strategies for therapeutics targeting the membrane of SARS-CoV-2 and other fusogenic viruses.
Collapse
Affiliation(s)
- David W Sanders
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Chanelle C Jumper
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Paul J Ackerman
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Dan Bracha
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Anita Donlic
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Hahn Kim
- Princeton University Small Molecule Screening Center, Princeton University, Princeton, United States
- Department of Chemistry, Princeton University, Princeton, United States
| | - Devin Kenney
- Department of Microbiology, Boston University School of Medicine, Boston, United States
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
| | - Ivan Castello-Serrano
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, United States
| | - Saori Suzuki
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Tomokazu Tamura
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Alexander H Tavares
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Mohsan Saeed
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, United States
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, United States
| | - Florian Douam
- Department of Microbiology, Boston University School of Medicine, Boston, United States
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, United States
| | - Robert F Padera
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States
| | - Clifford P Brangwynne
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
- Howard Hughes Medical Institute, Princeton, United States
| |
Collapse
|
164
|
Kluesner MG, Lahr WS, Lonetree CL, Smeester BA, Qiu X, Slipek NJ, Claudio Vázquez PN, Pitzen SP, Pomeroy EJ, Vignes MJ, Lee SC, Bingea SP, Andrew AA, Webber BR, Moriarity BS. CRISPR-Cas9 cytidine and adenosine base editing of splice-sites mediates highly-efficient disruption of proteins in primary and immortalized cells. Nat Commun 2021; 12:2437. [PMID: 33893286 PMCID: PMC8065034 DOI: 10.1038/s41467-021-22009-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 02/24/2021] [Indexed: 12/19/2022] Open
Abstract
CRISPR-Cas9 cytidine and adenosine base editors (CBEs and ABEs) can disrupt genes without introducing double-stranded breaks by inactivating splice sites (BE-splice) or by introducing premature stop (pmSTOP) codons. However, no in-depth comparison of these methods or a modular tool for designing BE-splice sgRNAs exists. To address these needs, we develop SpliceR ( http://z.umn.edu/spliceR ) to design and rank BE-splice sgRNAs for any Ensembl annotated genome, and compared disruption approaches in T cells using a screen against the TCR-CD3 MHC Class I immune synapse. Among the targeted genes, we find that targeting splice-donors is the most reliable disruption method, followed by targeting splice-acceptors, and introducing pmSTOPs. Further, the CBE BE4 is more effective for disruption than the ABE ABE7.10, however this disparity is eliminated by employing ABE8e. Collectively, we demonstrate a robust method for gene disruption, accompanied by a modular design tool that is of use to basic and translational researchers alike.
Collapse
Affiliation(s)
- Mitchell G Kluesner
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Walker S Lahr
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Cara-Lin Lonetree
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Branden A Smeester
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Xiaohong Qiu
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Nicholas J Slipek
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Patricia N Claudio Vázquez
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Samuel P Pitzen
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Emily J Pomeroy
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Madison J Vignes
- College of Biological Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Samantha C Lee
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
- College of Biological Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Samuel P Bingea
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Aneesha A Andrew
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
- College of Biological Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Beau R Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
| | - Branden S Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
165
|
Braun M, Aguilera AR, Sundarrajan A, Corvino D, Stannard K, Krumeich S, Das I, Lima LG, Meza Guzman LG, Li K, Li R, Salim N, Jorge MV, Ham S, Kelly G, Vari F, Lepletier A, Raghavendra A, Pearson S, Madore J, Jacquelin S, Effern M, Quine B, Koufariotis LT, Casey M, Nakamura K, Seo EY, Hölzel M, Geyer M, Kristiansen G, Taheri T, Ahern E, Hughes BGM, Wilmott JS, Long GV, Scolyer RA, Batstone MD, Landsberg J, Dietrich D, Pop OT, Flatz L, Dougall WC, Veillette A, Nicholson SE, Möller A, Johnston RJ, Martinet L, Smyth MJ, Bald T. CD155 on Tumor Cells Drives Resistance to Immunotherapy by Inducing the Degradation of the Activating Receptor CD226 in CD8 + T Cells. Immunity 2021; 53:805-823.e15. [PMID: 33053330 DOI: 10.1016/j.immuni.2020.09.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 05/21/2020] [Accepted: 09/15/2020] [Indexed: 12/20/2022]
Abstract
The activating receptor CD226 is expressed on lymphocytes, monocytes, and platelets and promotes anti-tumor immunity in pre-clinical models. Here, we examined the role of CD226 in the function of tumor-infiltrating lymphocytes (TILs) and resistance to immunotherapy. In murine tumors, a large proportion of CD8+ TILs had decreased surface expression of CD226 and exhibited features of dysfunction, whereas CD226hi TILs were highly functional. This correlation was seen also in TILs isolated from HNSCC patients. Mutation of CD226 at tyrosine 319 (Y319) led to increased CD226 surface expression, enhanced anti-tumor immunity and improved efficacy of immune checkpoint blockade (ICB). Mechanistically, tumor-derived CD155, the ligand for CD226, initiated phosphorylation of Y319 by Src kinases, thereby enabling ubiquitination of CD226 by CBL-B, internalization, and proteasomal degradation. In pre-treatment samples from melanoma patients, CD226+CD8+ T cells correlated with improved progression-free survival following ICB. Our findings argue for the development of therapies aimed at maintaining the expression of CD226.
Collapse
Affiliation(s)
- Matthias Braun
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Amelia Roman Aguilera
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Ashmitha Sundarrajan
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Dillon Corvino
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Kimberley Stannard
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Sophie Krumeich
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Indrajit Das
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Luize G Lima
- Tumor Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Lizeth G Meza Guzman
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Kunlun Li
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Rui Li
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, QC, Canada; Department of Medicine, McGill University, Montréal, QC, Canada
| | - Nazhifah Salim
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Maria Villancanas Jorge
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Sunyoung Ham
- Tumor Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Gabrielle Kelly
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Frank Vari
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Ailin Lepletier
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Ashwini Raghavendra
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Sally Pearson
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Jason Madore
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Sebastien Jacquelin
- Gordon and Jessie Gilmour Leukemia Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Maike Effern
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany; Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, VIC, Australia
| | - Brodie Quine
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Lambros T Koufariotis
- Medical Genomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Mika Casey
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Kyohei Nakamura
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Eun Y Seo
- Immuno-Oncology Discovery, Bristol-Myers Squibb, Redwood City, CA, USA
| | - Michael Hölzel
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Matthias Geyer
- Institute of Structural Biology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Glen Kristiansen
- Institute of Pathology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Touraj Taheri
- Pathology Queensland, Royal Brisbane and Women's Hospital, University of Queensland Herston, Herston, QLD, Australia
| | - Elizabeth Ahern
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; Royal Brisbane and Women's Hospital, University of Queensland Herston, Herston, QLD, Australia
| | - Brett G M Hughes
- Royal Brisbane and Women's Hospital, University of Queensland Herston, Herston, QLD, Australia
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; The University of Sydney, Central Clinical School, Sydney, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; The University of Sydney, Central Clinical School, Sydney, NSW, Australia; Royal North Shore Hospital, Sydney, NSW, Australia; Mater Hospital, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Martin D Batstone
- Royal Brisbane and Women's Hospital, University of Queensland Herston, Herston, QLD, Australia
| | - Jennifer Landsberg
- Department of Dermatology and Allergy, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Dimo Dietrich
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Oltin T Pop
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland; Department of Dermatology, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - William C Dougall
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - André Veillette
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, QC, Canada; Department of Medicine, McGill University, Montréal, QC, Canada; Department of Medicine, University of Montréal, Montréal, QC, Canada
| | - Sandra E Nicholson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Andreas Möller
- Tumor Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Robert J Johnston
- Immuno-Oncology Discovery, Bristol-Myers Squibb, Redwood City, CA, USA
| | - Ludovic Martinet
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse F-31000, France
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia.
| | - Tobias Bald
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia.
| |
Collapse
|
166
|
Gros OJ, Damstra HGJ, Kapitein LC, Akhmanova A, Berger F. Dynein self-organizes while translocating the centrosome in T-cells. Mol Biol Cell 2021; 32:855-868. [PMID: 33689395 PMCID: PMC8108531 DOI: 10.1091/mbc.e20-10-0668] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/12/2021] [Accepted: 03/04/2021] [Indexed: 12/16/2022] Open
Abstract
T-cells massively restructure their internal architecture upon reaching an antigen-presenting cell (APC) to form the immunological synapse (IS), a cell-cell interface necessary for efficient elimination of the APC. This reorganization occurs through tight coordination of cytoskeletal processes: actin forms a peripheral ring, and dynein motors translocate the centrosome toward the IS. A recent study proposed that centrosome translocation involves a microtubule (MT) bundle that connects the centrosome perpendicularly to dynein at the synapse center: the "stalk." The synapse center, however, is actin-depleted, while actin was assumed to anchor dynein. We propose that dynein is attached to mobile membrane anchors, and investigate this model with computer simulations. We find that dynein organizes into a cluster in the synapse when translocating the centrosome, aligning MTs into a stalk. By implementing both a MT-capture-shrinkage and a MT-sliding mechanism, we explicitly demonstrate that this organization occurs in both systems. However, results obtained with MT-sliding dynein are more robust and display a stalk morphology consistent with our experimental data obtained with expansion microscopy. Thus, our simulations suggest that actin organization in T-cells during activation defines a specific geometry in which MT-sliding dynein can self-organize into a cluster and cause stalk formation.
Collapse
Affiliation(s)
- Oane J Gros
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Hugo G J Damstra
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Florian Berger
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
167
|
Hammink R, Weiden J, Voerman D, Popelier C, Eggermont LJ, Schluck M, Figdor CG, Verdoes M. Semiflexible Immunobrushes Induce Enhanced T Cell Activation and Expansion. ACS APPLIED MATERIALS & INTERFACES 2021; 13:16007-16018. [PMID: 33797875 PMCID: PMC8045021 DOI: 10.1021/acsami.0c21994] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
A variety of bioactive materials developed to expand T cells for adoptive transfer into cancer patients are currently evaluated in the clinic. In most cases, T cell activating biomolecules are attached to rigid surfaces or matrices and form a static interface between materials and the signaling receptors on the T cells. We hypothesized that a T cell activating polymer brush interface might better mimic the cell surface of a natural antigen-presenting cell, facilitating receptor movement and concomitant advantageous mechanical forces to provide enhanced T cell activating capacities. Here, as a proof of concept, we synthesized semiflexible polyisocyanopeptide (PIC) polymer-based immunobrushes equipped with T cell activating agonistic anti-CD3 (αCD3) and αCD28 antibodies placed on magnetic microbeads. We demonstrated enhanced efficiency of ex vivo expansion of activated primary human T cells even at very low numbers of stimulating antibodies compared to rigid beads. Importantly, the immunobrush architecture appeared crucial for this improved T cell activating capacity. Immunobrushes outperform current benchmarks by producing higher numbers of T cells exhibiting a combination of beneficial phenotypic characteristics, such as reduced exhaustion marker expression, high cytokine production, and robust expression of cytotoxic hallmarks. This study indicates that semiflexible immunobrushes have great potential in making T cell-based immunotherapies more effective.
Collapse
Affiliation(s)
- Roel Hammink
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Jorieke Weiden
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Dion Voerman
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Carlijn Popelier
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
| | - Loek J. Eggermont
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Marjolein Schluck
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Carl G. Figdor
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Martijn Verdoes
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| |
Collapse
|
168
|
Tu H, Wu Z, Xia Y, Chen H, Hu H, Ding Z, Zhou F, Guo S. Profiling of immune-cancer interactions at the single-cell level using a microfluidic well array. Analyst 2021; 145:4138-4147. [PMID: 32409799 DOI: 10.1039/d0an00110d] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancer immunotherapy has achieved great success in hematological cancers. However, immune cells are a highly heterogeneous population and can vary highly in clonal expansion, migration and function status, making it difficult to evaluate and predict patient response to immune therapy. Conventional technologies only yield information on the average population information of the treatment, masking the heterogeneity of the individual T cell activation status, the formation of immune synapse, as well as the efficacy of tumor cell killing at the single-cell level. To fully interrogate these single-cell events in detail, herein, we present a microfluidic microwell array device that enables the massive parallel analysis of the immunocyte's heterogeneity upon its interaction pairs with tumor cells at the single-cell level. By precisely controlling the number and ratio of tumor cells and T cells, our technique can interrogate the dynamics of the CD8+ T cell and leukemia cell interaction inside 6400 microfluidic wells simultaneously. We have demonstrated that by investigating the interactions of T cell and tumor cell pairs at the single-cell level using our microfluidic chip, details hidden in bulk investigations, such as heterogeneity in T cell killing capacity, time-dependent killing dynamics, as well as drug treatment-induced dynamic shifts, can be revealed. This method opens up avenues to investigate the efficacy of cancer immunotherapy and resistance at the single-cell level and can explore our understanding of fundamental cancer immunity as well as determine cancer immunotherapy efficacy for personalized therapy.
Collapse
Affiliation(s)
- Honglei Tu
- Department of Clinical Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, People's Republic of China.
| | - Zhuhao Wu
- Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China.
| | - Yu Xia
- Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China.
| | - Hui Chen
- Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China.
| | - Hang Hu
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Zhao Ding
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Fuling Zhou
- Department of Clinical Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, People's Republic of China.
| | - Shishang Guo
- Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
169
|
Klebe M, Olbert PJ, Hofmann R, Barth PJ, Hegele A. [CpG-ODN instillation boosts ICAM-1 expression in an orthotopic murine UCC model: immunohistochemical evaluation of the local response to immunostimulatory DNA]. Aktuelle Urol 2021. [PMID: 33853160 DOI: 10.1055/a-1268-2069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Immunostimulatory CpG oligodeoxynucleotides (CpG-ODN) have been verified as an effective antineoplastic agent for intravesical application in a murine orthotopic C57-BL6 /MB-49 urothelial cell carcinoma (UCC). To date, many details in the mode of action have remained unclear. Preceding studies pointed towards a Th1-weighted response. The aim of this work was to identify the local lymphocyte subsets in murine tumour-bearing bladders and to examine effects on the expression of Intercellular Adhesion Molecule 1 (ICAM-1) after treatment with CpG-ODN. MATERIAL AND METHODS Different instillation schedules were applied in an established orthotopic C57-BL6 /MB49 UCC model. After 13 days, fresh frozen sections of the harvested bladders were immunohistochemically examined for the infiltration density of lymphocytes expressing CD 3, CD4, CD8 and CD19. In a second series of the same animal model, healthy and tumour-bearing bladders were exposed to CpG-ODN or PBS and later stained for the expression of ICAM-1. RESULTS CpG-ODN instillation led to augmented T-cell infiltration (represented by CD3). Further T-cell subdifferentiation between T-helper cells (CD4) and cytotoxic T cells (CD 8a) did not show a perceptible variety between groups. The B-cell population (CD19) was found to decrease over the course of treatment. In the second series, treatment provoked a strong expression of ICAM-1 by infiltrating leukocytes, endothelial cells and particularly by the cancer cells themselves. DISCUSSION The previously observed augmented lymphocyte density was classified as T-cell infiltration. The decline of the B-cell concentration over the course of treatment suggests a Th2 suppression in favour of a Th-1 polarisation. These findings support the assumption that a cell-mediated immune response is the mode of action underlying the antineoplastic CpG-ODN capacities. The marked upregulation of ICAM-1 expression, especially on tumour cells, suggests a crucial role of this membrane protein for the initiation and maintenance of anticancer immune response. CONCLUSION CpG-ODN might be a prospective alternative to established instillation therapies. With a view to the current BCG shortage and the well-known toxicities, an amplification of the topic therapy armamentarium could be achievable. The now described capability of ICAM-1 induction on carcinoma cells and, by association, the reversal of escape strategies to cancer immunity may also make the agent interesting as an adjuvant for modern checkpoint inhibition.
Collapse
Affiliation(s)
- Marwin Klebe
- Krankenhaus Nordwest, Klink für Urologie und Kinderurologie, Frankfurt
| | | | - Rainer Hofmann
- Universitätsklinikum Gießen und Marburg - Standort Marburg, Klinik für Urologie und Kinderurologie, Marburg
| | - Peter Josef Barth
- Universitätsklinikum Münster, Gerhard-Domagk-Institut für Pathologie, Münster
| | - Axel Hegele
- Urologisches Zentrum Mittelhessen, DRK-Krankenhaus Biedenkopf und Universitätsklinikum Gießen und Marburg, Klinik für Strahlentherapie, Standort Marburg
| |
Collapse
|
170
|
Kim HR, Park JS, Karabulut H, Yasmin F, Jun CD. Transgelin-2: A Double-Edged Sword in Immunity and Cancer Metastasis. Front Cell Dev Biol 2021; 9:606149. [PMID: 33898417 PMCID: PMC8060441 DOI: 10.3389/fcell.2021.606149] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
Transgelin-2, a small actin-binding protein, is the only transgelin family member expressed in immune cells. In T and B lymphocytes, transgelin-2 is constitutively expressed, but in antigen-presenting cells, it is significantly upregulated upon lipopolysaccharide stimulation. Transgelin-2 acts as a molecular staple to stabilize the actin cytoskeleton, and it competes with cofilin to bind filamentous (F)-actin. This action may enable immune synapse stabilization during T-cell interaction with cognate antigen-presenting cells. Furthermore, transgelin-2 blocks Arp2/3 complex-nucleated actin branching, which is presumably related to small filopodia formation, enhanced phagocytic function, and antigen presentation. Overall, transgelin-2 is an essential part of the molecular armament required for host defense against neoplasms and infectious diseases. However, transgelin-2 acts as a double-edged sword, as its expression is also essential for a wide range of tumor development, including drug resistance and metastasis. Thus, targeting transgelin-2 can also have a therapeutic advantage for cancer treatment; selectively suppressing transgelin-2 expression may prevent multidrug resistance in cancer chemotherapy. Here, we review newly discovered molecular characteristics of transgelin-2 and discuss clinical applications for cancer and immunotherapy.
Collapse
Affiliation(s)
- Hye-Ran Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea.,Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Jeong-Su Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea.,Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Hatice Karabulut
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea.,Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Fatima Yasmin
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea.,Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea.,Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| |
Collapse
|
171
|
Wabnitz GH, Honus S, Habicht J, Orlik C, Kirchgessner H, Samstag Y. LFA-1 cluster formation in T-cells depends on L-plastin phosphorylation regulated by P90 RSK and PP2A. Cell Mol Life Sci 2021; 78:3543-3564. [PMID: 33449151 PMCID: PMC11072591 DOI: 10.1007/s00018-020-03744-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 11/21/2020] [Accepted: 12/15/2020] [Indexed: 10/25/2022]
Abstract
The integrin LFA-1 is crucial for T-cell/ APC interactions and sensitive recognition of antigens. Precise nanoscale organization and valency regulation of LFA-1 are mandatory for an appropriate function of the immune system. While the inside-out signals regulating the LFA-1 affinity are well described, the molecular mechanisms controlling LFA-1 avidity are still not fully understood. Here, we show that activation of the actin-bundling protein L-plastin (LPL) through phosphorylation at serine-5 enables the formation of clusters containing LFA-1 in high-affinity conformation. Phosphorylation of LPL is induced by an nPKC-MEK-p90RSK pathway and counter-regulated by the serine-threonine phosphatase PP2A. Interestingly, recruitment of LFA-1 into the T-cell/APC contact zone is not affected by LPL phosphorylation. Instead, for this process, activation of the actin-remodeling protein cofilin through dephosphorylation is essential. Together, this study reveals a dichotomic spatial regulation of LFA-1 clustering and microscale movement in T-cells by two different actin-binding proteins, LPL and cofilin.
Collapse
Affiliation(s)
- Guido H Wabnitz
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany.
| | - Sibylle Honus
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Jüri Habicht
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Christian Orlik
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Henning Kirchgessner
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Yvonne Samstag
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| |
Collapse
|
172
|
CD101 as an indicator molecule for pathological changes at the interface of host-microbiota interactions. Int J Med Microbiol 2021; 311:151497. [PMID: 33773220 DOI: 10.1016/j.ijmm.2021.151497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/22/2021] [Accepted: 03/16/2021] [Indexed: 11/21/2022] Open
Abstract
Intestinal microbiota signal to local and distant tissues in the body. Thus, they also regulate biochemical, metabolic and immunological processes in the gut and in the pancreas. Vice versa, eating habits or the immune system of the host shape the intraluminal microbiota. Disruptions of these versatile host-microbiota interactions underlie the pathogenesis of complex immune-mediated disorders such as inflammatory bowel disease (IBD) or type 1 diabetes (T1D). Consequently, dysbiosis and increased intestinal permeability associated with both disorders change the biology of underlying tissues, as evidenced, for example, by an altered expression of surface markers such as CD101 on immune cells located at these dynamic host-microbiota interfaces. CD101, a heavily glycosylated member of the immunoglobulin superfamiliy, is predominantly detected on myeloid cells, intraepithelial lymphocytes (IELs) and regulatory T cells (Tregs) in the gut. The expression of CD101 on both myeloid cells and T lymphocytes protects from experimental enterocolitis and T1D. The improved outcome of both diseases is associated with an anti-inflammatory cytokine profile and a reduced expansion of T cells. However, distinct bacteria suppress the expression of CD101 on myeloid cells, similar as does inflammation on T cells. Thus, the reduced CD101 expression in T1D and IBD patients might be a consequence of an altered composition of the intestinal microbiota, enhanced bacterial translocation and a subsequent primining of local and systemic inflammatory immune responses.
Collapse
|
173
|
Dawson KA, Yan Y. Current understanding of biological identity at the nanoscale and future prospects. NATURE NANOTECHNOLOGY 2021; 16:229-242. [PMID: 33597736 DOI: 10.1038/s41565-021-00860-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/19/2021] [Indexed: 06/12/2023]
Abstract
Nanoscale objects are processed by living organisms using highly evolved and sophisticated endogenous cellular networks, specifically designed to manage objects of this size. While these processes potentially allow nanostructures unique access to and control over key biological machineries, they are also highly protected by cell or host defence mechanisms at all levels. A thorough understanding of bionanoscale recognition events, including the molecules involved in the cell recognition machinery, the nature of information transferred during recognition processes and the coupled downstream cellular processing, would allow us to achieve a qualitatively novel form of biological control and advanced therapeutics. Here we discuss evolving fundamental microscopic and mechanistic understanding of biological nanoscale recognition. We consider the interface between a nanostructure and a target cell membrane, outlining the categories of nanostructure properties that are recognized, and the associated nanoscale signal transduction and cellular programming mechanisms that constitute biological recognition.
Collapse
Affiliation(s)
- Kenneth A Dawson
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Dublin, Ireland.
| | - Yan Yan
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Dublin, Ireland.
- School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
174
|
Gunasinghe SD, Peres NG, Goyette J, Gaus K. Biomechanics of T Cell Dysfunctions in Chronic Diseases. Front Immunol 2021; 12:600829. [PMID: 33717081 PMCID: PMC7948521 DOI: 10.3389/fimmu.2021.600829] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Understanding the mechanisms behind T cell dysfunctions during chronic diseases is critical in developing effective immunotherapies. As demonstrated by several animal models and human studies, T cell dysfunctions are induced during chronic diseases, spanning from infections to cancer. Although factors governing the onset and the extent of the functional impairment of T cells can differ during infections and cancer, most dysfunctional phenotypes share common phenotypic traits in their immune receptor and biophysical landscape. Through the latest developments in biophysical techniques applied to explore cell membrane and receptor-ligand dynamics, we are able to dissect and gain further insights into the driving mechanisms behind T cell dysfunctions. These insights may prove useful in developing immunotherapies aimed at reinvigorating our immune system to fight off infections and malignancies more effectively. The recent success with checkpoint inhibitors in treating cancer opens new avenues to develop more effective, targeted immunotherapies. Here, we highlight the studies focused on the transformation of the biophysical landscape during infections and cancer, and how T cell biomechanics shaped the immunopathology associated with chronic diseases.
Collapse
Affiliation(s)
- Sachith D Gunasinghe
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Newton G Peres
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Jesse Goyette
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
175
|
Ben-Shmuel A, Sabag B, Biber G, Barda-Saad M. The Role of the Cytoskeleton in Regulating the Natural Killer Cell Immune Response in Health and Disease: From Signaling Dynamics to Function. Front Cell Dev Biol 2021; 9:609532. [PMID: 33598461 PMCID: PMC7882700 DOI: 10.3389/fcell.2021.609532] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/11/2021] [Indexed: 01/13/2023] Open
Abstract
Natural killer (NK) cells are innate lymphoid cells, which play key roles in elimination of virally infected and malignant cells. The balance between activating and inhibitory signals derived from NK surface receptors govern the NK cell immune response. The cytoskeleton facilitates most NK cell effector functions, such as motility, infiltration, conjugation with target cells, immunological synapse assembly, and cytotoxicity. Though many studies have characterized signaling pathways that promote actin reorganization in immune cells, it is not completely clear how particular cytoskeletal architectures at the immunological synapse promote effector functions, and how cytoskeletal dynamics impact downstream signaling pathways and activation. Moreover, pioneering studies employing advanced imaging techniques have only begun to uncover the architectural complexity dictating the NK cell activation threshold; it is becoming clear that a distinct organization of the cytoskeleton and signaling receptors at the NK immunological synapse plays a decisive role in activation and tolerance. Here, we review the roles of the actin cytoskeleton in NK cells. We focus on how actin dynamics impact cytolytic granule secretion, NK cell motility, and NK cell infiltration through tissues into inflammatory sites. We will also describe the additional cytoskeletal components, non-muscle Myosin II and microtubules that play pivotal roles in NK cell activity. Furthermore, special emphasis will be placed on the role of the cytoskeleton in assembly of immunological synapses, and how mutations or downregulation of cytoskeletal accessory proteins impact NK cell function in health and disease.
Collapse
Affiliation(s)
- Aviad Ben-Shmuel
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Batel Sabag
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Guy Biber
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Mira Barda-Saad
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
176
|
Schoppmeyer R, van Buul JD. The diapedesis synapse: dynamic leukocyte-endothelium interactions. CURRENT OPINION IN PHYSIOLOGY 2021. [DOI: 10.1016/j.cophys.2020.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
177
|
Kim HR, Park JS, Fatima Y, Kausar M, Park JH, Jun CD. Potentiating the Antitumor Activity of Cytotoxic T Cells via the Transmembrane Domain of IGSF4 That Increases TCR Avidity. Front Immunol 2021; 11:591054. [PMID: 33597944 PMCID: PMC7882689 DOI: 10.3389/fimmu.2020.591054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/14/2020] [Indexed: 01/25/2023] Open
Abstract
A robust T-cell response is an important component of sustained antitumor immunity. In this respect, the avidity of TCR in the antigen-targeting of tumors is crucial for the quality of the T-cell response. This study reports that the transmembrane (TM) domain of immunoglobulin superfamily member 4 (IGSF4) binds to the TM of the CD3 ζ-chain through an interaction between His177 and Asp36, which results in IGSF4-CD3 ζ dimers. IGSF4 also forms homo-dimers through the GxxVA motif in the TM domain, thereby constituting large TCR clusters. Overexpression of IGSF4 lacking the extracellular (IG4ΔEXT) domain potentiates the OTI CD8+ T cells to release IFN-γ and TNF-α and to kill OVA+-B16F10 melanoma cells. In animal models, IG4ΔEXT significantly reduces B16F10 tumor metastasis as well as tumor growth. Collectively, the results indicate that the TM domain of IGSF4 can regulate TCR avidity, and they further demonstrate that TCR avidity regulation is critical for improving the antitumor activity of cytotoxic T cells.
Collapse
MESH Headings
- Animals
- Cell Adhesion Molecule-1/genetics
- Cell Adhesion Molecule-1/immunology
- Cell Line, Tumor
- Humans
- Immunotherapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice, Inbred C57BL
- Mice, Transgenic
- Protein Domains
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- T-Lymphocytes/immunology
- Mice
Collapse
Affiliation(s)
- Hye-Ran Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Jeong-Su Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Yasmin Fatima
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Maiza Kausar
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Jin-Hwa Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| |
Collapse
|
178
|
McKay ZP, Brown MC, Gromeier M. Aryl Hydrocarbon Receptor Signaling Controls CD155 Expression on Macrophages and Mediates Tumor Immunosuppression. THE JOURNAL OF IMMUNOLOGY 2021; 206:1385-1394. [PMID: 33504618 DOI: 10.4049/jimmunol.2000792] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 01/05/2021] [Indexed: 01/02/2023]
Abstract
Crosstalk between costimulatory and coinhibitory ligands are a prominent node of immune cell regulation. Mounting evidence points toward a critical role for CD155, the poliovirus receptor, in suppressing T cell function, particularly in cancer. However, relative to other known costimulatory/coinhibitory ligands (e.g., CD86, CD80, PD-L1), the physiological functions of CD155 and the mechanisms controlling its expression remain unclear. We discovered that CD155 expression is coregulated with PD-L1 on tumor-associated macrophages, is transcriptionally regulated by persistently active aryl hydrocarbon receptor (AhR), and can be targeted for suppression via AhR inhibition in vivo. Therapeutic inhibition of AhR reversed tumor immunosuppression in an immune competent murine tumor model, and markers of AhR activity were highly correlated with tumor-associated macrophage markers in human glioblastomas. Thus, CD155 functions within a broader, AhR-controlled macrophage activation phenotype that can be targeted to reverse tumor immunosuppression.
Collapse
Affiliation(s)
- Zachary P McKay
- Department of Neurosurgery, Duke University Medical School, Durham, NC 27710
| | - Michael C Brown
- Department of Neurosurgery, Duke University Medical School, Durham, NC 27710
| | - Matthias Gromeier
- Department of Neurosurgery, Duke University Medical School, Durham, NC 27710
| |
Collapse
|
179
|
Felce JH, Parolini L, Sezgin E, Céspedes PF, Korobchevskaya K, Jones M, Peng Y, Dong T, Fritzsche M, Aarts D, Frater J, Dustin ML. Single-Molecule, Super-Resolution, and Functional Analysis of G Protein-Coupled Receptor Behavior Within the T Cell Immunological Synapse. Front Cell Dev Biol 2021; 8:608484. [PMID: 33537301 PMCID: PMC7848080 DOI: 10.3389/fcell.2020.608484] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/21/2020] [Indexed: 12/31/2022] Open
Abstract
A central process in immunity is the activation of T cells through interaction of T cell receptors (TCRs) with agonistic peptide-major histocompatibility complexes (pMHC) on the surface of antigen presenting cells (APCs). TCR-pMHC binding triggers the formation of an extensive contact between the two cells termed the immunological synapse, which acts as a platform for integration of multiple signals determining cellular outcomes, including those from multiple co-stimulatory/inhibitory receptors. Contributors to this include a number of chemokine receptors, notably CXC-chemokine receptor 4 (CXCR4), and other members of the G protein-coupled receptor (GPCR) family. Although best characterized as mediators of ligand-dependent chemotaxis, some chemokine receptors are also recruited to the synapse and contribute to signaling in the absence of ligation. How these and other GPCRs integrate within the dynamic structure of the synapse is unknown, as is how their normally migratory Gαi-coupled signaling is terminated upon recruitment. Here, we report the spatiotemporal organization of several GPCRs, focusing on CXCR4, and the G protein Gαi2 within the synapse of primary human CD4+ T cells on supported lipid bilayers, using standard- and super-resolution fluorescence microscopy. We find that CXCR4 undergoes orchestrated phases of reorganization, culminating in recruitment to the TCR-enriched center. This appears to be dependent on CXCR4 ubiquitination, and does not involve stable interactions with TCR microclusters, as viewed at the nanoscale. Disruption of this process by mutation impairs CXCR4 contributions to cellular activation. Gαi2 undergoes active exclusion from the synapse, partitioning from centrally-accumulated CXCR4. Using a CRISPR-Cas9 knockout screen, we identify several diverse GPCRs with contributions to T cell activation, most significantly the sphingosine-1-phosphate receptor S1PR1, and the oxysterol receptor GPR183. These, and other GPCRs, undergo organization similar to CXCR4; including initial exclusion, centripetal transport, and lack of receptor-TCR interactions. These constitute the first observations of GPCR dynamics within the synapse, and give insights into how these receptors may contribute to T cell activation. The observation of broad GPCR contributions to T cell activation also opens the possibility that modulating GPCR expression in response to cell status or environment may directly regulate responsiveness to pMHC.
Collapse
Affiliation(s)
- James H Felce
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Lucia Parolini
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Erdinc Sezgin
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Pablo F Céspedes
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | | | - Mathew Jones
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Yanchun Peng
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Tao Dong
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Marco Fritzsche
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom.,Rosalind Franklin Institute, Didcot, United Kingdom
| | - Dirk Aarts
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, United Kingdom
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,National Institute of Health Research Biomedical Research Centre, Oxford, United Kingdom
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
180
|
Cremasco F, Menietti E, Speziale D, Sam J, Sammicheli S, Richard M, Varol A, Klein C, Umana P, Bacac M, Colombetti S, Perro M. Cross-linking of T cell to B cell lymphoma by the T cell bispecific antibody CD20-TCB induces IFNγ/CXCL10-dependent peripheral T cell recruitment in humanized murine model. PLoS One 2021; 16:e0241091. [PMID: 33406104 PMCID: PMC7787458 DOI: 10.1371/journal.pone.0241091] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022] Open
Abstract
Diffuse large B cell lymphomas (DLBCL) are a highly heterogeneous subtype of Non Hodgkin Lymphoma (NHL), accounting for about 25% of NHL. Despite an increased progression-free survival upon therapy, 40-50% of patients develop relapse/refractory disease, therefore there remains an important medical need. T cell recruiting therapies, such as the CD20xCD3 T cell bi-specific antibody CD20-TCB (RG6026 or glofitamab), represent a novel approach to target all stages of DLBCL, especially those that fail to respond to multiple lines of treatment. We aimed for a better understanding of the molecular features related to the mode of action (MoA) of CD20-TCB in inducing Target/T cell synapse formation and human T cell recruitment to the tumor. To directly evaluate the correlation between synapse, cytokine production and anti-tumor efficacy using CD20-TCB, we developed an innovative preclinical human DLBCL in vivo model that allowed tracking in vivo human T cell dynamics by multiphoton intravital microscopy (MP-IVM). By ex vivo and in vivo approaches, we revealed that CD20-TCB is inducing strong and stable synapses between human T cell and tumor cells, which are dependent on the dose of CD20-TCB and on LFA-1 activity but not on FAS-L. Moreover, despite CD20-TCB being a large molecule (194.342 kDa), we observed that intra-tumor CD20-TCB-mediated human T cell-tumor cell synapses occur within 1 hour upon CD20-TCB administration. These tight interactions, observed for at least 72 hours post TCB administration, result in tumor cell cytotoxicity, resident T cell proliferation and peripheral blood T cell recruitment into tumor. By blocking the IFNγ-CXCL10 axis, the recruitment of peripheral T cells was abrogated, partially affecting the efficacy of CD20-TCB treatment which rely only on resident T cell proliferation. Altogether these data reveal that CD20-TCB's anti-tumor activity relies on a triple effect: i) fast formation of stable T cell-tumor cell synapses which induce tumor cytotoxicity and cytokine production, ii) resident T cell proliferation and iii) recruitment of fresh peripheral T cells to the tumor core to allow a positive enhancement of the anti-tumor effect.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/pharmacology
- Antigens, CD20/immunology
- Antineoplastic Agents, Immunological/pharmacology
- Cell Line, Tumor
- Chemokine CXCL10/immunology
- Humans
- Interferon-gamma/immunology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Mice
- Neoplasm Proteins/immunology
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
| | | | | | - Johannes Sam
- Roche Innovation Center Zürich, Zürich, Switzerland
| | | | | | - Ahmet Varol
- Roche Innovation Center Zürich, Zürich, Switzerland
| | | | - Pablo Umana
- Roche Innovation Center Zürich, Zürich, Switzerland
| | - Marina Bacac
- Roche Innovation Center Zürich, Zürich, Switzerland
| | | | - Mario Perro
- Roche Innovation Center Zürich, Zürich, Switzerland
- * E-mail:
| |
Collapse
|
181
|
Wardell CM, MacDonald KN, Levings MK, Cook L. Cross talk between human regulatory T cells and antigen-presenting cells: Lessons for clinical applications. Eur J Immunol 2020; 51:27-38. [PMID: 33301176 DOI: 10.1002/eji.202048746] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/04/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022]
Abstract
Regulatory T cells (Tregs) have a critical role in maintaining self-tolerance and immune homeostasis. There is much interest in using Tregs as a cell therapy to re-establish tolerance in conditions such as inflammatory bowel disease and type 1 diabetes, with many ongoing clinical studies testing the safety and efficacy of this approach. Manufacturing of Tregs for therapy typically involves ex vivo expansion to obtain sufficient cell numbers for infusion and comes with the risk of altering the activity of key biological processes. However, this process also offers an opportunity to tailor Treg function to maximize in vivo activity. In this review, we focus on the roles of antigen-presenting cells (APCs) in the generation and function of Tregs in humans. In addition to stimulating the development of Tregs, APCs activate Tregs and provide signals that induce specialized functional and homing marker expression. Cross talk between Tregs and APCs is a critical, often under-appreciated, aspect of Treg biology, with APCs mediating the key properties of infectious tolerance and bystander suppression. Understanding the biology of human Treg-APC interactions will reveal new ways to optimize Treg-based therapeutic approaches.
Collapse
Affiliation(s)
- Christine M Wardell
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Katherine N MacDonald
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Laura Cook
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
182
|
Muntjewerff EM, Meesters LD, van den Bogaart G, Revelo NH. Reverse Signaling by MHC-I Molecules in Immune and Non-Immune Cell Types. Front Immunol 2020; 11:605958. [PMID: 33384693 PMCID: PMC7770133 DOI: 10.3389/fimmu.2020.605958] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/13/2020] [Indexed: 12/16/2022] Open
Abstract
Major histocompatibility complex (MHC) molecules are well-known for their role in antigen (cross-) presentation, thereby functioning as key players in the communication between immune cells, for example dendritic cells (DCs) and T cells, or immune cells and their targets, such as T cells and virus-infected or tumor cells. However, much less appreciated is the fact that MHC molecules can also act as signaling receptors. In this process, here referred to as reverse MHC class I (MHC-I) signaling, ligation of MHC molecules can lead to signal-transduction and cell regulatory effects in the antigen presenting cell. In the case of MHC-I, reverse signaling can have several outcomes, including apoptosis, migration, induced or reduced proliferation and cytotoxicity towards target cells. Here, we provide an overview of studies showing the signaling pathways and cell outcomes upon MHC-I stimulation in various immune and non-immune cells. Signaling molecules like RAC-alpha serine/threonine-protein kinase (Akt1), extracellular signal-regulated kinases 1/2 (ERK1/2), and nuclear factor-κB (NF-κB) were common signaling molecules activated upon MHC-I ligation in multiple cell types. For endothelial and smooth muscle cells, the in vivo relevance of reverse MHC-I signaling has been established, namely in the context of adverse effects after tissue transplantation. For other cell types, the role of reverse MHC-I signaling is less clear, since aspects like the in vivo relevance, natural MHC-I ligands and the extended downstream pathways are not fully known.The existing evidence, however, suggests that reverse MHC-I signaling is involved in the regulation of the defense against bacterial and viral infections and against malignancies. Thereby, reverse MHC-I signaling is a potential target for therapies against viral and bacterial infections, cancer immunotherapies and management of organ transplantation outcomes.
Collapse
Affiliation(s)
- Elke M Muntjewerff
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Luca D Meesters
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Molecular Microbiology and Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| | - Natalia H Revelo
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
183
|
Castro-Sanchez P, Teagle AR, Prade S, Zamoyska R. Modulation of TCR Signaling by Tyrosine Phosphatases: From Autoimmunity to Immunotherapy. Front Cell Dev Biol 2020; 8:608747. [PMID: 33425916 PMCID: PMC7793860 DOI: 10.3389/fcell.2020.608747] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Early TCR signaling is dependent on rapid phosphorylation and dephosphorylation of multiple signaling and adaptor proteins, leading to T cell activation. This process is tightly regulated by an intricate web of interactions between kinases and phosphatases. A number of tyrosine phosphatases have been shown to modulate T cell responses and thus alter T cell fate by negatively regulating early TCR signaling. Mutations in some of these enzymes are associated with enhanced predisposition to autoimmunity in humans, and mouse models deficient in orthologous genes often show T cell hyper-activation. Therefore, phosphatases are emerging as potential targets in situations where it is desirable to enhance T cell responses, such as immune responses to tumors. In this review, we summarize the current knowledge about tyrosine phosphatases that regulate early TCR signaling and discuss their involvement in autoimmunity and their potential as targets for tumor immunotherapy.
Collapse
Affiliation(s)
- Patricia Castro-Sanchez
- Ashworth Laboratories, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Alexandra R Teagle
- Ashworth Laboratories, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Sonja Prade
- Ashworth Laboratories, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Rose Zamoyska
- Ashworth Laboratories, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
184
|
Bein J, Thurner L, Hansmann M, Hartmann S. Lymphocyte predominant cells of nodular lymphocyte predominant Hodgkin lymphoma interact with rosetting T cells in an immunological synapse. Am J Hematol 2020; 95:1495-1502. [PMID: 32815561 DOI: 10.1002/ajh.25972] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/05/2020] [Accepted: 08/17/2020] [Indexed: 12/21/2022]
Abstract
Nodular lymphocyte predominant Hodgkin lymphoma (NLPHL) is a subtype of Hodgkin lymphoma with a preserved B-cell phenotype and follicular T helper (TFH ) cells rosetting around the tumor cells, the lymphocyte-predominant (LP) cells. As we recently described reactivity of the B-cell receptors of LP cells of some NLPHL cases with Moraxella spp. proteins, we hypothesized that LP cells could present peptides to rosetting T cells in a major histocompatibility complex class II (MHCII)-bound manner. Rosetting PD1+ T cells were present in the majority of NLPHL cases, both in typical (17/20) and variant patterns (16/19). In most cases, T-cell rosettes were CD69+ (typical NLPHL, 17/20; NLPHL variant, 14/19). Furthermore, both MHCII alpha and beta chains were expressed in the LP cells in 23/39 NLPHL. Proximity ligation assay and confocal laser imaging demonstrated interaction of the MHCII beta chain expressed by the LP cells and the T-cell receptor alpha chain expressed by rosetting T cells. We thus conclude that rosetting T cells in NLPHL express markers that are encountered after antigenic exposure, that MHCII is expressed by the LP cells, and that LP cells interact with rosetting T cells in an immunological synapse in a subset of cases. As they likely receive growth stimulatory signals in this way, blockade of this interaction, for example, by PD1-directed checkpoint inhibitors, could be a treatment option in a subset of cases in the future.
Collapse
Affiliation(s)
- Julia Bein
- Dr. Senckenberg Institute of Pathology Goethe University Frankfurt am Main Germany
| | - Lorenz Thurner
- Department of Internal Medicine I and José Carreras Center for Immuno‐ and Gene Therapy Saarland University Medical School Homburg/Saar Germany
| | - Martin‐Leo Hansmann
- Dr. Senckenberg Institute of Pathology Goethe University Frankfurt am Main Germany
- Frankfurt Institute of Advanced Studies Frankfurt am Main Germany
| | - Sylvia Hartmann
- Dr. Senckenberg Institute of Pathology Goethe University Frankfurt am Main Germany
| |
Collapse
|
185
|
Lühr JJ, Alex N, Amon L, Kräter M, Kubánková M, Sezgin E, Lehmann CHK, Heger L, Heidkamp GF, Smith AS, Zaburdaev V, Böckmann RA, Levental I, Dustin ML, Eggeling C, Guck J, Dudziak D. Maturation of Monocyte-Derived DCs Leads to Increased Cellular Stiffness, Higher Membrane Fluidity, and Changed Lipid Composition. Front Immunol 2020; 11:590121. [PMID: 33329576 PMCID: PMC7728921 DOI: 10.3389/fimmu.2020.590121] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/15/2020] [Indexed: 01/02/2023] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells of the immune system. Upon sensing pathogenic material in their environment, DCs start to mature, which includes cellular processes, such as antigen uptake, processing and presentation, as well as upregulation of costimulatory molecules and cytokine secretion. During maturation, DCs detach from peripheral tissues, migrate to the nearest lymph node, and find their way into the correct position in the net of the lymph node microenvironment to meet and interact with the respective T cells. We hypothesize that the maturation of DCs is well prepared and optimized leading to processes that alter various cellular characteristics from mechanics and metabolism to membrane properties. Here, we investigated the mechanical properties of monocyte-derived dendritic cells (moDCs) using real-time deformability cytometry to measure cytoskeletal changes and found that mature moDCs were stiffer compared to immature moDCs. These cellular changes likely play an important role in the processes of cell migration and T cell activation. As lipids constitute the building blocks of the plasma membrane, which, during maturation, need to adapt to the environment for migration and DC-T cell interaction, we performed an unbiased high-throughput lipidomics screening to identify the lipidome of moDCs. These analyses revealed that the overall lipid composition was significantly changed during moDC maturation, even implying an increase of storage lipids and differences of the relative abundance of membrane lipids upon maturation. Further, metadata analyses demonstrated that lipid changes were associated with the serum low-density lipoprotein (LDL) and cholesterol levels in the blood of the donors. Finally, using lipid packing imaging we found that the membrane of mature moDCs revealed a higher fluidity compared to immature moDCs. This comprehensive and quantitative characterization of maturation associated changes in moDCs sets the stage for improving their use in clinical application.
Collapse
Affiliation(s)
- Jennifer J. Lühr
- Laboratory of Dendritic Cell Biology, Department of Dermatology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
- Nano-Optics, Max-Planck Institute for the Science of Light, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Nils Alex
- Department of Physics, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Martin Kräter
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Biological Optomechanics, Max-Planck Institute for the Science of Light, Erlangen, Germany
| | - Markéta Kubánková
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Biological Optomechanics, Max-Planck Institute for the Science of Light, Erlangen, Germany
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Raddcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Christian H. K. Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Gordon F. Heidkamp
- Laboratory of Dendritic Cell Biology, Department of Dermatology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, pRED, Munich, Germany
| | - Ana-Sunčana Smith
- PULS Group, Department of Physics, IZNF, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Vasily Zaburdaev
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Mathematics in Life Sciences, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
- Medical Immunology Campus Erlangen, Erlangen, Germany
| | - Rainer A. Böckmann
- Computational Biology, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ilya Levental
- McGovern Medical School, The University of Texas Health Science Center, Houston, TX, United States
| | - Michael L. Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Christian Eggeling
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Raddcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Institute for Applied Optics and Biophysics, Friedrich-Schiller University Jena, Jena, Germany
- Leibniz Institute of Photonic Technologies e.V., Jena, Germany
| | - Jochen Guck
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Biological Optomechanics, Max-Planck Institute for the Science of Light, Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
- Medical Immunology Campus Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
| |
Collapse
|
186
|
Gómez-Henao W, Tenorio EP, Sanchez FRC, Mendoza MC, Ledezma RL, Zenteno E. Relevance of glycans in the interaction between T lymphocyte and the antigen presenting cell. Int Rev Immunol 2020; 40:274-288. [PMID: 33205679 DOI: 10.1080/08830185.2020.1845331] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The immunological synapse promotes receptors and ligands interaction in the contact interface between the T lymphocyte and the antigen presenting cell; glycosylation of the proteins involved in this biological process favors regulation of molecular interactions and development of the T lymphocyte effector response. Glycans in the immunological synapse influence cellular and molecular processes such as folding, expression, and structural stability of proteins, they also mediate ligand-receptor interaction and propagation of the intracellular signaling or inhibition of uncontrolled cellular activation that could lead to the development of autoimmunity, among others. It has been suggested that altered glycosylation of proteins that participate in the immunological synapse affects the signaling processes and cell proliferation, as well as exacerbation of the effector mechanisms of T cells that trigger systemic damage and autoimmunity. Understanding the role of glycans in the immune response has allowed for advances in the development of immunotherapies in different fields through the controlled and specific activation of the immune response. This review describes the structural and biological aspects of glycans associated with some molecules present in the immunological synapse, providing information that allows understanding the function of glycosylation in the interaction between the T lymphocyte and the antigen-presenting cell, as well as its impact on signaling and development regulation of T lymphocytes effector response.
Collapse
Affiliation(s)
- Wilton Gómez-Henao
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan; Mexico.,Cell Growth, Tissue Repair and Regeneration (CRRET), CNRS ERL 9215, Université Paris Est Créteil (UPEC), Créteil, France
| | - Eda Patricia Tenorio
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan; Mexico
| | | | - Miguel Cuéllar Mendoza
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan; Mexico
| | - Ricardo Lascurain Ledezma
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan; Mexico
| | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan; Mexico
| |
Collapse
|
187
|
Simões IT, Aranda F, Casadó-Llombart S, Velasco-de Andrés M, Català C, Álvarez P, Consuegra-Fernández M, Orta-Mascaró M, Merino R, Merino J, Alberola-Ila J, González-Aseguinolaza G, Carreras E, Martínez V, Lozano F. Multifaceted effects of soluble human CD6 in experimental cancer models. J Immunother Cancer 2020; 8:jitc-2019-000172. [PMID: 32217757 PMCID: PMC7174071 DOI: 10.1136/jitc-2019-000172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2020] [Indexed: 12/11/2022] Open
Abstract
Background CD6 is a lymphocyte surface co-receptor physically associated with the T-cell receptor (TCR)/CD3 complex at the center of the immunological synapse. There, CD6 assists in cell-to-cell contact stabilization and modulation of activation/differentiation events through interaction with CD166/ALCAM (activated leukocyte cell adhesion molecule), its main reported ligand. While accumulating evidence is attracting new interest on targeting CD6 for therapeutic purposes in autoimmune disorders, little is known on its potential in cancer. In an attempt to elucidate the in vivo relevance of blocking CD6-mediated interactions in health and disease, we explored the consequences of expressing high circulating levels of a soluble form CD6 (sCD6) as a decoy receptor. Methods High sCD6 serum levels were achieved by using transgenic C57BL/6 mice expressing human sCD6 under the control of lymphoid-specific transcriptional elements (shCD6LckEμTg) or wild type either transduced with hepatotropic adeno-associated virus coding for mouse sCD6 or undergoing repeated infusions of recombinant human sCD6 protein. Characterization of sCD6-induced changes was performed by ex vivo flow cytometry and functional analyses of mouse lymphoid organ cells. The in vivo relevance of those changes was explored by challenging mice with subcutaneous or metastatic tumors induced by syngeneic cancer cells of different lineage origins. Results Through a combination of in vitro and in vivo studies, we show that circulating sCD6 expression induces defective regulatory T cell (Treg) generation and function, decreased CD166/ALCAM-mediated tumor cell proliferation/migration and impaired galectin-induced T-cell apoptosis, supporting the fact that sCD6 modulates antitumor lymphocyte effector function and tumorigenesis. Accordingly, sCD6 expression in vivo resulted in delayed subcutaneous tumor growth and/or reduced metastasis on challenge of mice with syngeneic cancer cells. Conclusions Evidence is provided for the disruption of CD6 receptor–ligand interactions as a feasible immunomodulatory approach in cancer.
Collapse
Affiliation(s)
- Inês T Simões
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Fernando Aranda
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Sergi Casadó-Llombart
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - María Velasco-de Andrés
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Cristina Català
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Pilar Álvarez
- Departamento de Biología Molecular, Universidad de Cantabria-IDIVAL, Santander, Cantabria, Spain
| | - Marta Consuegra-Fernández
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Marc Orta-Mascaró
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Ramón Merino
- Instituto de Biomedicina y Biotecnología de Cantabria, CSIC-UC, Santander, Cantabria, Spain
| | - Jesús Merino
- Departamento de Biología Molecular, Universidad de Cantabria-IDIVAL, Santander, Cantabria, Spain
| | - José Alberola-Ila
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | | | - Esther Carreras
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Vanesa Martínez
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Francisco Lozano
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain .,Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Barcelona, Barcelona, Spain.,Servei d'Immunologia, Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
188
|
Varlet E, Ovejero S, Martinez AM, Cavalli G, Moreaux J. Role of Polycomb Complexes in Normal and Malignant Plasma Cells. Int J Mol Sci 2020; 21:ijms21218047. [PMID: 33126754 PMCID: PMC7662980 DOI: 10.3390/ijms21218047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 02/01/2023] Open
Abstract
Plasma cells (PC) are the main effectors of adaptive immunity, responsible for producing antibodies to defend the body against pathogens. They are the result of a complex highly regulated cell differentiation process, taking place in several anatomical locations and involving unique genetic events. Pathologically, PC can undergo tumorigenesis and cause a group of diseases known as plasma cell dyscrasias, including multiple myeloma (MM). MM is a severe disease with poor prognosis that is characterized by the accumulation of malignant PC within the bone marrow, as well as high clinical and molecular heterogeneity. MM patients frequently develop resistance to treatment, leading to relapse. Polycomb group (PcG) proteins are epigenetic regulators involved in cell fate and carcinogenesis. The emerging roles of PcG in PC differentiation and myelomagenesis position them as potential therapeutic targets in MM. Here, we focus on the roles of PcG proteins in normal and malignant plasma cells, as well as their therapeutic implications.
Collapse
Affiliation(s)
- Emmanuel Varlet
- Institute of Human Genetics, UMR 9002 Centre National de la Recherche Scientifique, University of Montpellier, Montpellier, 34396 Montpellier, France; (E.V.); (S.O.); (A.-M.M.); (G.C.)
| | - Sara Ovejero
- Institute of Human Genetics, UMR 9002 Centre National de la Recherche Scientifique, University of Montpellier, Montpellier, 34396 Montpellier, France; (E.V.); (S.O.); (A.-M.M.); (G.C.)
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France
| | - Anne-Marie Martinez
- Institute of Human Genetics, UMR 9002 Centre National de la Recherche Scientifique, University of Montpellier, Montpellier, 34396 Montpellier, France; (E.V.); (S.O.); (A.-M.M.); (G.C.)
| | - Giacomo Cavalli
- Institute of Human Genetics, UMR 9002 Centre National de la Recherche Scientifique, University of Montpellier, Montpellier, 34396 Montpellier, France; (E.V.); (S.O.); (A.-M.M.); (G.C.)
| | - Jerome Moreaux
- Institute of Human Genetics, UMR 9002 Centre National de la Recherche Scientifique, University of Montpellier, Montpellier, 34396 Montpellier, France; (E.V.); (S.O.); (A.-M.M.); (G.C.)
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France
- UFR Medicine, University of Montpellier, 34003 Montpellier, France
- Institut Universitaire de France (IUF), 75005 Paris, France
- Correspondence: ; Tel.: +33-04-6733-7903
| |
Collapse
|
189
|
Borowicz P, Chan H, Hauge A, Spurkland A. Adaptor proteins: Flexible and dynamic modulators of immune cell signalling. Scand J Immunol 2020; 92:e12951. [DOI: 10.1111/sji.12951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/22/2020] [Accepted: 07/26/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Paweł Borowicz
- Department of Molecular Medicine Institute of Basic Medical Sciences University of Oslo Oslo Norway
| | - Hanna Chan
- Department of Molecular Medicine Institute of Basic Medical Sciences University of Oslo Oslo Norway
| | - Anette Hauge
- Department of Molecular Medicine Institute of Basic Medical Sciences University of Oslo Oslo Norway
| | - Anne Spurkland
- Department of Molecular Medicine Institute of Basic Medical Sciences University of Oslo Oslo Norway
| |
Collapse
|
190
|
Rukhlenko O, Kholodenko BN, Kolch W. Systems biology approaches to macromolecules: the role of dynamic protein assemblies in information processing. Curr Opin Struct Biol 2020; 67:61-68. [PMID: 33126139 DOI: 10.1016/j.sbi.2020.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/10/2020] [Accepted: 09/16/2020] [Indexed: 11/17/2022]
Abstract
Macromolecular protein assemblies govern many cellular processes and are disturbed in many diseases including cancer. Often seen as static molecular machines, protein complexes involved in signal transduction networks exhibit intricate dynamics that are critical for their function. Using the RAS-RAF-MEK-ERK pathway as example we discuss recent progress in our understanding of protein complex dynamics achieved through mathematical modelling, computational simulations and structural studies. The emerging picture highlights that both spatial and temporal dynamics cooperate to enable correct signal processing and the fine tuning of timing, duration and strengths of signalling. These dynamic processes are subverted by oncogenic mutations and contribute to tumorigenesis and drug resistance.
Collapse
Affiliation(s)
- Oleksii Rukhlenko
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Boris N Kholodenko
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland; Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland; Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
191
|
Abstract
Sentrin/small ubiquitin-like modifier (SUMO) is protein modification pathway that regulates multiple biological processes, including cell division, DNA replication/repair, signal transduction, and cellular metabolism. In this review, we will focus on recent advances in the mechanisms of disease pathogenesis, such as cancer, diabetes, seizure, and heart failure, which have been linked to the SUMO pathway. SUMO is conjugated to lysine residues in target proteins through an isopeptide linkage catalyzed by SUMO-specific activating (E1), conjugating (E2), and ligating (E3) enzymes. In steady state, the quantity of SUMO-modified substrates is usually a small fraction of unmodified substrates due to the deconjugation activity of the family Sentrin/SUMO-specific proteases (SENPs). In contrast to the complexity of the ubiquitination/deubiquitination machinery, the biochemistry of SUMOylation and de-SUMOylation is relatively modest. Specificity of the SUMO pathway is achieved through redox regulation, acetylation, phosphorylation, or other posttranslational protein modification of the SUMOylation and de-SUMOylation enzymes. There are three major SUMOs. SUMO-1 usually modifies a substrate as a monomer; however, SUMO-2/3 can form poly-SUMO chains. The monomeric SUMO-1 or poly-SUMO chains can interact with other proteins through SUMO-interactive motif (SIM). Thus SUMO modification provides a platform to enhance protein-protein interaction. The consequence of SUMOylation includes changes in cellular localization, protein activity, or protein stability. Furthermore, SUMO may join force with ubiquitin to degrade proteins through SUMO-targeted ubiquitin ligases (STUbL). After 20 yr of research, SUMO has been shown to play critical roles in most, if not all, biological pathways. Thus the SUMO enzymes could be targets for drug development to treat human diseases.
Collapse
Affiliation(s)
- Hui-Ming Chang
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Edward T H Yeh
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
192
|
Janssens I, Cools N. Regulating the regulators: Is introduction of an antigen-specific approach in regulatory T cells the next step to treat autoimmunity? Cell Immunol 2020; 358:104236. [PMID: 33137651 DOI: 10.1016/j.cellimm.2020.104236] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/28/2020] [Accepted: 10/04/2020] [Indexed: 12/22/2022]
Abstract
In autoimmunity, the important and fragile balance between immunity and tolerance is disturbed, resulting in abnormal immune responses to the body's own tissues and cells. CD4+CD25hiFoxP3+ regulatory T cells (Tregs) induce peripheral tolerance in vivo by means of direct cell-cell contact and release of soluble factors, or indirectly through antigen-presenting cells (APC), thereby controlling auto-reactive effector T cells. Based on these unique capacities of Tregs, preclinical studies delivered proof-of-principle for the clinical use of Tregs for the treatment of autoimmune diseases. To date, the first clinical trials using ex vivo expanded polyclonal Tregs have been completed. These pioneering studies demonstrate the feasibility of generating large numbers of polyclonal Tregs in a good manufacturing practices (GMP)-compliant manner, and that infusion of Tregs is well tolerated by patients with no evidence of general immunosuppression. Nonetheless, only modest clinical results were observed, arguing that a more antigen-specific approach might be needed to foster a durable patient-specific clinical cell therapy without the risk for general immunosuppression. In this review, we discuss current knowledge, applications and future goals of adoptive immune-modulatory Treg therapy for the treatment of autoimmune disease and transplant rejection. We describe the key advances and prospects of the potential use of T cell receptor (TCR)- and chimeric antigen receptor (CAR)-engineered Tregs in future clinical applications. These approaches could deliver the long-awaited breakthrough in stopping undesired autoimmune responses and transplant rejections.
Collapse
Affiliation(s)
- Ibo Janssens
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium.
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
193
|
Chandler NJ, Call MJ, Call ME. T Cell Activation Machinery: Form and Function in Natural and Engineered Immune Receptors. Int J Mol Sci 2020; 21:E7424. [PMID: 33050044 PMCID: PMC7582382 DOI: 10.3390/ijms21197424] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/15/2022] Open
Abstract
The impressive success of chimeric antigen receptor (CAR)-T cell therapies in treating advanced B-cell malignancies has spurred a frenzy of activity aimed at developing CAR-T therapies for other cancers, particularly solid tumors, and optimizing engineered T cells for maximum clinical benefit in many different disease contexts. A rapidly growing body of design work is examining every modular component of traditional single-chain CARs as well as expanding out into many new and innovative engineered immunoreceptor designs that depart from this template. New approaches to immune cell and receptor engineering are being reported with rapidly increasing frequency, and many recent high-quality reviews (including one in this special issue) provide comprehensive coverage of the history and current state of the art in CAR-T and related cellular immunotherapies. In this review, we step back to examine our current understanding of the structure-function relationships in natural and engineered lymphocyte-activating receptors, with an eye towards evaluating how well the current-generation CAR designs recapitulate the most desirable features of their natural counterparts. We identify key areas that we believe are under-studied and therefore represent opportunities to further improve our grasp of form and function in natural and engineered receptors and to rationally design better therapeutics.
Collapse
Affiliation(s)
- Nicholas J. Chandler
- Structural Biology Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; (N.J.C.); (M.J.C.)
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Melissa J. Call
- Structural Biology Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; (N.J.C.); (M.J.C.)
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Matthew E. Call
- Structural Biology Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; (N.J.C.); (M.J.C.)
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
194
|
Baxter AA. Stoking the Fire: How Dying Cells Propagate Inflammatory Signalling through Extracellular Vesicle Trafficking. Int J Mol Sci 2020; 21:ijms21197256. [PMID: 33019535 PMCID: PMC7583891 DOI: 10.3390/ijms21197256] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023] Open
Abstract
Communication between dying cells and their environment is a critical process that promotes tissue homeostasis during normal cellular turnover, whilst during disease settings, it can contribute to inflammation through the release of intracellular factors. Extracellular vesicles (EVs) are a heterogeneous class of membrane-bound cell-derived structures that can engage in intercellular communication via the trafficking of bioactive molecules between cells and tissues. In addition to the well-described functions of EVs derived from living cells, the ability of dying cells to release EVs capable of mediating functions on target cells or tissues is also of significant interest. In particular, during inflammatory settings such as acute tissue injury, infection and autoimmunity, the EV-mediated transfer of proinflammatory cargo from dying cells is an important process that can elicit profound proinflammatory effects in recipient cells and tissues. Furthermore, the biogenesis of EVs via unique cell-death-associated pathways has also been recently described, highlighting an emerging niche in EV biology. This review outlines the mechanisms and functions of dying-cell-derived EVs and their ability to drive inflammation during various modes of cell death, whilst reflecting on the challenges and knowledge gaps in investigating this subgenre of extracellular vesicles research.
Collapse
Affiliation(s)
- Amy A Baxter
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| |
Collapse
|
195
|
Qin W, Chen L, Wang Z, Li Q, Fan C, Wu M, Zhang Y. Bioinspired DNA Nanointerface with Anisotropic Aptamers for Accurate Capture of Circulating Tumor Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000647. [PMID: 33042737 PMCID: PMC7539197 DOI: 10.1002/advs.202000647] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/29/2020] [Indexed: 05/08/2023]
Abstract
The capture and analysis of circulating tumor cells (CTCs) have provided a non-invasive entry for cancer diagnosis and disease monitoring. Despite recent development in affinity-based CTCs isolation, it remains challenging to achieve efficient capture toward CTCs with dynamic surface expression. Enlightened by the synergistic effect insideimmune synapses, the development of a nanointerface engineered with topology-defined anisotropic aptamers programmed by DNA scaffold (DNA nanosynapse), for accurate CTCs isolation, is herein reported. As compared to isotropic aptamers, the DNA nanosynapse exhibits enhanced anchoring on the cell membrane with both high and low epithelial cell adhesion molecule (EpCAM) expression. This nanointerface enables accurate capture toward CTCs of heterogeneous EpCAM, without dramatically proportional change inside the mixture of diverse phenotypes. By applying this nanoplatform, CTCs detection as well as downstream analysis for measuring disease status can be achieved in clinical samples from breast cancer patients.
Collapse
Affiliation(s)
- Weiwei Qin
- Guangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhouGuangdong510006China
- College of Materials and EnergySouth China Agricultural UniversityGuangzhouGuangdong510642China
- State Key Laboratory of Chemo/Biosensing and ChemometricsHunan UniversityChangsha410082China
| | - Liang Chen
- Guangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhouGuangdong510006China
| | - Zhiru Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhouGuangdong510006China
| | - Qian Li
- School of Chemistry and Chemical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Chunhai Fan
- School of Chemistry and Chemical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Minhao Wu
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
| | - Yuanqing Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhouGuangdong510006China
| |
Collapse
|
196
|
Carpenter MA, Wang Y, Telmer CA, Schmidt BF, Yang Z, Bruchez MP. Protein Proximity Observed Using Fluorogen Activating Protein and Dye Activated by Proximal Anchoring (FAP-DAPA) System. ACS Chem Biol 2020; 15:2433-2443. [PMID: 32786268 DOI: 10.1021/acschembio.0c00419] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The development and function of tissues, blood, and the immune system is dependent upon proximity for cellular recognition and communication. However, the detection of cell-to-cell contacts is limited due to a lack of reversible, quantitative probes that can function at these dynamic sites of irregular geometry. Described here is a novel chemo-genetic tool developed for fluorescent detection of protein-protein proximity and cell apposition that utilizes the Fluorogen Activating Protein (FAP) in combination with a Dye Activated by Proximal Anchoring (DAPA). The FAP-DAPA system has two protein components, the HaloTag and FAP, expressed on separate protein targets or in separate cells. The proteins function to bind and activate a compound that has the hexyl chloride (HexCl) ligand connected to malachite green (MG), the FAP fluorogen, via a poly(ethylene glycol) spacer spanning up to 28 nm. The dehalogenase protein, HaloTag, covalently binds the HexCl ligand, locally concentrating the attached MG. If the FAP is within range of the anchored fluorogen, it will bind and activate MG specifically when the bath concentration is too low to saturate the FAP receptor. A new FAP variant was isolated with a 1000-fold reduced KD of ∼10-100 nM so that the fluorogen activation reports proximity without artificially enhancing it. The system was characterized using purified FRB and FKBP fusion proteins and showed a doubling of fluorescence upon rapamycin induced complex formation. In cocultured HEK293 cells (HaloTag and FAP-expressing) fluorescence increased at contact sites across a broad range of labeling conditions, more reliably providing contact-specific fluorescence activation with the lower-affinity FAP variant. When combined with suitable targeting and expression constructs, this labeling system may offer significant improvements in on-demand detection of intercellular contacts, potentially applicable in neurological and immunological synapse measurements and other transient, dynamic biological appositions that can be perturbed using other labeling methods that stabilize these interactions.
Collapse
Affiliation(s)
- M. Alexandra Carpenter
- Carnegie Mellon University, Department of Chemistry, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Yi Wang
- Carnegie Mellon University, Department of Biological Sciences, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Cheryl A. Telmer
- Carnegie Mellon University, Department of Biological Sciences, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States
- Carnegie Mellon University, Molecular Biosensor and Imaging Center, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Brigitte F. Schmidt
- Carnegie Mellon University, Molecular Biosensor and Imaging Center, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Zhipeng Yang
- Carnegie Mellon University, Department of Biological Sciences, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Marcel P. Bruchez
- Carnegie Mellon University, Department of Chemistry, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States
- Carnegie Mellon University, Department of Biological Sciences, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States
- Carnegie Mellon University, Molecular Biosensor and Imaging Center, 5000 Forbes Avenue, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
197
|
Raskov H, Orhan A, Christensen JP, Gögenur I. Cytotoxic CD8 + T cells in cancer and cancer immunotherapy. Br J Cancer 2020; 124:359-367. [PMID: 32929195 PMCID: PMC7853123 DOI: 10.1038/s41416-020-01048-4] [Citation(s) in RCA: 709] [Impact Index Per Article: 177.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/15/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022] Open
Abstract
The functions of, and interactions between, the innate and adaptive immune systems are vital for anticancer immunity. Cytotoxic T cells expressing cell-surface CD8 are the most powerful effectors in the anticancer immune response and form the backbone of current successful cancer immunotherapies. Immune-checkpoint inhibitors are designed to target immune-inhibitory receptors that function to regulate the immune response, whereas adoptive cell-transfer therapies use CD8+ T cells with genetically modified receptors—chimaeric antigen receptors—to specify and enhance CD8+ T-cell functionality. New generations of cytotoxic T cells with genetically modified or synthetic receptors are being developed and evaluated in clinical trials. Furthermore, combinatory regimens might optimise treatment effects and reduce adverse events. This review summarises advances in research on the most prominent immune effectors in cancer and cancer immunotherapy, cytotoxic T cells, and discusses possible implications for future cancer treatment.
Collapse
Affiliation(s)
- Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark.
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
198
|
Macpherson AM, Barry SC, Ricciardelli C, Oehler MK. Epithelial Ovarian Cancer and the Immune System: Biology, Interactions, Challenges and Potential Advances for Immunotherapy. J Clin Med 2020; 9:E2967. [PMID: 32937961 PMCID: PMC7564553 DOI: 10.3390/jcm9092967] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/28/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022] Open
Abstract
Recent advances in the understanding of immune function and the interactions with tumour cells have led to the development of various cancer immunotherapies and strategies for specific cancer types. However, despite some stunning successes with some malignancies such as melanomas and lung cancer, most patients receive little or no benefit from immunotherapy, which has been attributed to the tumour microenvironment and immune evasion. Although the US Food and Drug Administration have approved immunotherapies for some cancers, to date, only the anti-angiogenic antibody bevacizumab is approved for the treatment of epithelial ovarian cancer. Immunotherapeutic strategies for ovarian cancer are still under development and being tested in numerous clinical trials. A detailed understanding of the interactions between cancer and the immune system is vital for optimisation of immunotherapies either alone or when combined with chemotherapy and other therapies. This article, in two main parts, provides an overview of: (1) components of the normal immune system and current knowledge regarding tumour immunology, biology and their interactions; (2) strategies, and targets, together with challenges and potential innovative approaches for cancer immunotherapy, with attention given to epithelial ovarian cancer.
Collapse
Affiliation(s)
- Anne M. Macpherson
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
| | - Simon C. Barry
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide 5005, Australia;
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
| | - Martin K. Oehler
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide 5000, Australia
| |
Collapse
|
199
|
Watkins TS, Miles JJ. The human T-cell receptor repertoire in health and disease and potential for omics integration. Immunol Cell Biol 2020; 99:135-145. [PMID: 32677130 DOI: 10.1111/imcb.12377] [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: 05/14/2020] [Revised: 07/07/2020] [Accepted: 07/12/2020] [Indexed: 12/11/2022]
Abstract
The adaptive immune system arose 600 million years ago in a cold-blooded fish. Over countless generations, our antecedents tuned the function of the T-cell receptor (TCR). The TCR system is arguably the most complex known to science. The TCR evolved hypervariability to fight the hypervariability of pathogens and cancers that look to consume our resources. This review describes the genetics and architecture of the human TCR and highlights surprising new discoveries over the past years that have disproved very old dogmas. The standardization of TCR sequencing data is discussed in preparation for big data bioinformatics and predictive analysis. We next catalogue new signatures and phenomenon discovered by TCR next generation sequencing (NGS) in health and disease and work that remain to be done in this space. Finally, we discuss how TCR NGS can add to immunodiagnostics and integrate with other omics platforms for both a deeper understanding of TCR biology and its use in the clinical setting.
Collapse
Affiliation(s)
- Thomas S Watkins
- The Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Cairns, QLD, Australia.,Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia.,Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
| | - John J Miles
- The Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Cairns, QLD, Australia.,Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia.,Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
200
|
Correlation of host inflammatory cytokines and immune-related metabolites, but not viral NS1 protein, with disease severity of dengue virus infection. PLoS One 2020; 15:e0237141. [PMID: 32764789 PMCID: PMC7413495 DOI: 10.1371/journal.pone.0237141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 07/21/2020] [Indexed: 12/13/2022] Open
Abstract
Severe dengue can be lethal caused by manifestations such as severe bleeding, fluid accumulation and organ impairment. This study aimed to investigate the role of dengue non-structural 1 (NS1) protein and host factors contributing to severe dengue. Electrical cell-substrate impedance sensing system was used to investigate the changes in barrier function of microvascular endothelial cells treated NS1 protein and serum samples from patients with different disease severity. Cytokines and metabolites profiles were assessed using a multiplex cytokine assay and liquid chromatography mass spectrometry respectively. The findings showed that NS1 was able to induce the loss of barrier function in microvascular endothelium in a dose dependent manner, however, the level of NS1 in serum samples did not correlate with the extent of vascular leakage induced. Further assessment of host factors revealed that cytokines such as CCL2, CCL5, CCL20 and CXCL1, as well as adhesion molecule ICAM-1, that are involved in leukocytes infiltration were expressed higher in dengue patients in comparison to healthy individuals. In addition, metabolomics study revealed the presence of deregulated metabolites involved in the phospholipid metabolism pathway in patients with severe manifestations. In conclusion, disease severity in dengue virus infection did not correlate directly with NS1 level, but instead with host factors that are involved in the regulation of junctional integrity and phospholipid metabolism. However, as the studied population was relatively small in this study, these exploratory findings should be confirmed by expanding the sample size using an independent cohort to further establish the significance of this study.
Collapse
|