1
|
Lui Y, Ferreira Fernandes J, Vuong MT, Sharma S, Santos AM, Davis SJ. The Structural Biology of T-Cell Antigen Detection at Close Contacts. Immunol Rev 2025; 331:e70014. [PMID: 40181535 PMCID: PMC11969063 DOI: 10.1111/imr.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 02/16/2025] [Indexed: 04/05/2025]
Abstract
T cells physically interrogate their targets using tiny membrane protrusions called microvilli, forming junctions ~400 nm in diameter and ~ 15 nm deep, referred to as "close contacts". These contacts, which are stabilized by the binding of the small adhesion protein CD2 to its ligand, CD58 and locally exclude large proteins such as the phosphatase CD45, are the sites of antigen recognition by the T-cell receptor (TCR) and very early signaling by T cells. With our collaborators, we have characterized the molecular structures of several of the key proteins mediating these early events: i.e., CD2 and its ligands, CD45, the αβ- and γδ-TCRs, and the accessory proteins CD28, CTLA-4, and PD-1. Here, we review our structural work and the insights it offers into the early events underpinning T-cell responsiveness that take place in the confined space of the close contact. We reflect on the crucial roles that the structural organization and dimensions of these proteins are likely to have in determining the sequence of events leading to antigen recognition at close contacts and consider the general implications of the structural work for explanations of how immune receptor signaling is initiated.
Collapse
Affiliation(s)
- Yuan Lui
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - João Ferreira Fernandes
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Mai T. Vuong
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Sumana Sharma
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Ana Mafalda Santos
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Simon J. Davis
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| |
Collapse
|
2
|
Borowska MT, Liu LD, Caveney NA, Jude KM, Kim WJ, Masubuchi T, Hui E, Majzner RG, Garcia KC. Orientation-dependent CD45 inhibition with viral and engineered ligands. Sci Immunol 2024; 9:eadp0707. [PMID: 39454026 PMCID: PMC11537708 DOI: 10.1126/sciimmunol.adp0707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/01/2024] [Indexed: 10/27/2024]
Abstract
CD45 is a cell surface phosphatase that shapes the T cell receptor signaling threshold but does not have a known ligand. A family of adenovirus proteins, including E3/49K, exploits CD45 to evade immunity by binding to the extracellular domain of CD45, resulting in the suppression of T cell signaling. We determined the cryo-EM structure of this complex and found that the E3/49K protein is composed of three immunoglobulin domains assembled as "beads on a string" that compel CD45 into a closely abutted dimer by cross-linking the CD45 D3 domain, leading to steric inhibition of its intracellular phosphatase activity. Inspired by the E3/49K mechanism, we engineered CD45 surrogate ligands that can fine-tune T cell activation by dimerizing CD45 into different orientations and proximities. The adenovirus E3/49K protein has taught us that, despite a lack of a known ligand, CD45 activity can be modulated by extracellular dimerizing ligands that perturb its phosphatase activity and alter T cell responses.
Collapse
Affiliation(s)
- Marta T. Borowska
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94063, USA
| | - Liu D. Liu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94063, USA
| | - Nathanael A. Caveney
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94063, USA
| | - Kevin M. Jude
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94063, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94063, USA
| | - Won-Ju Kim
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Takeya Masubuchi
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Enfu Hui
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robbie G. Majzner
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - K. Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94063, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94063, USA
- Department of Structural Biology, Stanford University, Stanford, CA 94063, USA
| |
Collapse
|
3
|
Hildenbrand A, Cramer P, Bertolotti M, Kaiser NS, Kläsener K, Nickel CM, Reth M, Heim A, Hengel H, Burgert HG, Ruzsics Z. Inhibition of B cell receptor signaling induced by the human adenovirus species D E3/49K protein. Front Immunol 2024; 15:1432226. [PMID: 39139562 PMCID: PMC11321000 DOI: 10.3389/fimmu.2024.1432226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction The early transcription unit 3 (E3) of human adenoviruses (HAdVs) encodes several immunoevasins, including the E3/49K protein, which is unique for species D of HAdVs. It is expressed as surface transmembrane protein and shed. E3/49K of HAdV-D64 binds to the protein tyrosine phosphatase surface receptor CD45, thereby modulating activation of T and NK cells. Methods Considering that E3/49K represents the most polymorphic viral protein among species D HAdVs, we demonstrate here that all tested E3/49K orthologs bind to the immunologically important regulator CD45. Thus, this feature is conserved regardless of the pathological associations of the respective HAdV types. Results It appeared that modulation of CD45 is a unique property restricted to HAdVs of species D. Moreover, E3/49K treatment inhibited B cell receptor (BCR) signaling and impaired BCR signal phenotypes. The latter were highly comparable to B cells having defects in the expression of CD45, suggesting E3/49K as a potential tool to investigate CD45 specific functions. Conclusion We identified B cells as new direct target of E3/49K-mediated immune modulation, representing a novel viral immunosubversive mechanism.
Collapse
Affiliation(s)
- Andreas Hildenbrand
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Precious Cramer
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Milena Bertolotti
- Signaling Research Centers CIBSS and BIOSS, University of Freiburg, Freiburg, Germany
- Navita S.r.l., University of Eastern Piedmont A. Avogadro, Novara, Italy
| | - Nathalie Sophia Kaiser
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kathrin Kläsener
- Department of Rheumatology and Clinical Immunology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Clara Muriel Nickel
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael Reth
- Signaling Research Centers CIBSS and BIOSS, University of Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Albert Heim
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Hartmut Hengel
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans-Gerhard Burgert
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Zsolt Ruzsics
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
4
|
Abstract
T cell activation is initiated by the recognition of specific antigenic peptides and subsequently accomplished by complex signaling cascades. These aspects have been extensively studied for decades as pivotal factors in the establishment of adaptive immunity. However, how receptors or signaling molecules are organized in the resting state prior to encountering antigens has received less attention. Recent advancements in super-resolution microscopy techniques have revealed topographically controlled pre-formed organization of key molecules involved in antigen recognition and signal transduction on microvillar projections of T cells before activation and substantial effort has been dedicated to characterizing the topological structure of resting T cells over the past decade. This review will summarize our current understanding of how key surface receptors are pre-organized on the T-cell plasma membrane and discuss the potential role of these receptors, which are preassembled prior to ligand binding in the early activation events of T cells.
Collapse
Affiliation(s)
- Yunmin Jung
- Department of Nano-Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science, Seoul, Republic of Korea
| |
Collapse
|
5
|
Windheim M, Reubold TF, Aichane K, Gaestel M, Burgert HG. Enforced Dimerization of CD45 by the Adenovirus E3/49K Protein Inhibits T Cell Receptor Signaling. J Virol 2023; 97:e0189822. [PMID: 37125921 PMCID: PMC10231199 DOI: 10.1128/jvi.01898-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/07/2023] [Indexed: 05/02/2023] Open
Abstract
Human adenoviruses (HAdVs) are widespread pathogens that generally cause mild infections in immunocompetent individuals but severe or even fatal diseases in immunocompromised patients. In order to counteract the host immune defenses, HAdVs encode various immunomodulatory proteins in the early transcription unit 3 (E3). The E3/49K protein is a highly glycosylated type I transmembrane protein uniquely expressed by species D HAdVs. Its N-terminal ectodomain sec49K is released by metalloprotease-mediated shedding at the cell surface and binds to the receptor-like protein tyrosine phosphatase CD45, a critical regulator of leukocyte activation and functions. It remained elusive which domains of CD45 and E3/49K are involved in the interaction and whether such an interaction can also occur on the cell surface with membrane-anchored full-length E3/49K. Here, we show that the two extracellular domains R1 and R2 of E3/49K bind to the same site in the domain d3 of CD45. This interaction enforces the dimerization of CD45, causing the inhibition of T cell receptor signaling. Intriguingly, the membrane-anchored E3/49K appears to be designed like a "molecular fishing rod" using an extended disordered region of E3/49K as a "fishing line" to bridge the distance between the plasma membrane of infected cells and the CD45 binding site on T cells to effectively position the domains R1 and R2 as baits for CD45 binding. This design strongly suggests that both secreted sec49K as well as membrane-anchored full-length E3/49K have immunomodulatory functions. The forced dimerization of CD45 may be applied as a therapeutic strategy in chronic inflammatory disorders and cancer. IMPORTANCE The battle between viruses and their hosts is an ongoing arms race. Whereas the host tries to detect and eliminate the virus, the latter counteracts such antiviral measures to replicate and spread. Adenoviruses have evolved various mechanisms to evade the human immune response. The E3/49K protein of species D adenoviruses mediates the inhibition of immune cell function via binding to the protein tyrosine phosphatase CD45. Here, we show that E3/49K triggers the dimerization of CD45 and thereby inhibits its phosphatase activity. Intriguingly, the membrane-anchored E3/49K seems to be designed like a "molecular fishing rod" with the two CD45 binding domains of E3/49K as baits positioned at the end of an extended disordered region reminiscent of a fishing line. The adenoviral strategy to inhibit CD45 activity by forced dimerization may be used for therapeutic intervention in autoimmune diseases or to prevent graft rejection after transplantation.
Collapse
Affiliation(s)
- Mark Windheim
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Thomas F. Reubold
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Khadija Aichane
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Hans-Gerhard Burgert
- Institute of Virology, University Medical Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| |
Collapse
|
6
|
Koretzky GA. Building on the Past, Meeting the Moment. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:849-854. [PMID: 36947823 DOI: 10.4049/jimmunol.2390003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Affiliation(s)
- Gary A Koretzky
- Department of Internal Medicine, Weill Cornell Medicine, New York, NY
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca NY
| |
Collapse
|
7
|
Tetraspanin CD53 controls T cell immunity through regulation of CD45RO stability, mobility, and function. Cell Rep 2022; 39:111006. [PMID: 35767951 DOI: 10.1016/j.celrep.2022.111006] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 05/02/2022] [Accepted: 06/03/2022] [Indexed: 11/22/2022] Open
Abstract
T cells depend on the phosphatase CD45 to initiate T cell receptor signaling. Although the critical role of CD45 in T cells is established, the mechanisms controlling function and localization in the membrane are not well understood. Moreover, the regulation of specific CD45 isoforms in T cell signaling remains unresolved. By using unbiased mass spectrometry, we identify the tetraspanin CD53 as a partner of CD45 and show that CD53 controls CD45 function and T cell activation. CD53-negative T cells (Cd53-/-) exhibit substantial proliferation defects, and Cd53-/- mice show impaired tumor rejection and reduced IFNγ-producing T cells compared with wild-type mice. Investigation into the mechanism reveals that CD53 is required for CD45RO expression and mobility. In addition, CD53 is shown to stabilize CD45 on the membrane and is required for optimal phosphatase activity and subsequent Lck activation. Together, our findings reveal CD53 as a regulator of CD45 activity required for T cell immunity.
Collapse
|
8
|
She W, Shao J, Jia R. Targeting Splicing Factor SRSF6 for Cancer Therapy. Front Cell Dev Biol 2021; 9:780023. [PMID: 34917618 PMCID: PMC8669609 DOI: 10.3389/fcell.2021.780023] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/12/2021] [Indexed: 01/19/2023] Open
Abstract
Aberrant alternative splicing of pre-mRNA is an emerging cancer hallmark. Many cancer-associated genes undergo alternative splicing to produce multiple isoforms with diverse or even antagonistic functions. Oncogenic isoforms are often up-regulated, whereas tumor suppressive isoforms are down-regulated during tumorigenesis. Serine/arginine-rich splicing factor 6 (SRSF6) is an important splicing factor that regulates the alternative splicing of hundreds of target genes, including many cancer-associated genes. The potential roles of SRSF6 in cancers have attracted increasing attentions in the past decade. Accumulated pieces of evidence have shown that SRSF6 is a potential oncogenic gene that promotes oncogenic splicing when overexpressed. Targeting SRSF6 may suppress tumorigenesis. In this review, we describe the gene, mRNA, and protein structure of SRSF6; summarize the current understanding of the expression, functions, and regulatory mechanisms of SRSF6 during tumorigenesis; and discuss the potential application of targeting SRSF6 in cancer treatment.
Collapse
Affiliation(s)
- Wenting She
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Jun Shao
- Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology and Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan, China
| | - Rong Jia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Abstract
Like all herpesviruses, the roseoloviruses (HHV6A, -6B, and -7) establish lifelong infection within their host, requiring these viruses to evade host antiviral responses. One common host-evasion strategy is the downregulation of host-encoded, surface-expressed glycoproteins. Roseoloviruses have been shown to evade the host immune response by downregulating NK-activating ligands, class I MHC, and the TCR/CD3 complex. To more globally identify glycoproteins that are differentially expressed on the surface of HHV6A-infected cells, we performed cell surface capture of N-linked glycoproteins present on the surface of T cells infected with HHV6A, and compared these to proteins present on the surface of uninfected T cells. We found that the protein tyrosine phosphatase CD45 is downregulated in T cells infected with HHV6A. We also demonstrated that CD45 is similarly downregulated in cells infected with HHV7. CD45 is essential for signaling through the T cell receptor and, as such, is necessary for developing a fully functional immune response. Interestingly, the closely related betaherpesviruses human cytomegalovirus (HCMV) and murine cytomegalovirus (MCMV) have also separately evolved unique mechanisms to target CD45. While HCMV and MCMV target CD45 signaling and trafficking, HHV6A acts to downregulate CD45 transcripts. IMPORTANCE Human herpesviruses-6 and -7 infect essentially 100% of the world's population before the age of 5 and then remain latent or persistent in their host throughout life. As such, these viruses are among the most pervasive and stealthy of all viruses. Host immune cells rely on the presence of surface-expressed proteins to identify and target virus-infected cells. Here, we investigated the changes that occur to proteins expressed on the cell surface of T cells after infection with human herpesvirus-6A. We discovered that HHV-6A infection results in a reduction of CD45 on the surface of infected T cells and impaired activation in response to T cell receptor stimulation.
Collapse
|
10
|
Bhatta A, Chan MA, Benedict SH. Engagement of CD45 alters early signaling events in human T cells co-stimulated through TCR + CD28. Cell Immunol 2020; 353:104130. [PMID: 32446033 DOI: 10.1016/j.cellimm.2020.104130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/03/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
Previously our lab has shown that co-stimulation of human T cells through different co-stimulatory molecules tune differentiation to different phenotypes. An open question is where in the signaling pathways induced by the co-stimulation do differences occur that contribute to outcome of differentiation. In this project, we investigate the early signaling process by comparing events that follow engagement of CD45 alone or in association with a co-stimulatory molecule: CD28. CD45 plays a crucial role to initiate T cell signaling by dephosphorylating a negatively regulatory tyrosine residue in Src family kinases such as Lck. First, we observed that engagement of CD45 alone induced signaling in T cells. We observed that TCR/CD3 stimulation with CD45 promoted prolonged Lck association with TCR/CD3 complex and Lck remained associated during TCR/CD3 + CD28 + CD45 stimulation as well. We concluded that Lck association is dependent on TCR/CD3 and CD45 engagement. Hence, CD45 altered early signaling events in T cells.
Collapse
Affiliation(s)
- Anuja Bhatta
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, United States.
| | - Marcia A Chan
- Department of Pediatrics, Division of Allergy, Asthma, and Immunology, Children's Mercy Hospital, Kansas City, MO, United States
| | - Stephen H Benedict
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
11
|
Vignard V, Labbé M, Marec N, André-Grégoire G, Jouand N, Fonteneau JF, Labarrière N, Fradin D. MicroRNAs in Tumor Exosomes Drive Immune Escape in Melanoma. Cancer Immunol Res 2019; 8:255-267. [PMID: 31857348 DOI: 10.1158/2326-6066.cir-19-0522] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 10/14/2019] [Accepted: 12/10/2019] [Indexed: 11/16/2022]
Abstract
MicroRNAs (miRNA), small noncoding RNAs that regulate gene expression, exist not only in cells but also in a variety of body fluids. These circulating miRNAs could enable intercellular communication. miRNAs are packaged in membrane-encapsulated vesicles, such as exosomes, or protected by RNA-binding proteins. Here, we report that miRNAs included in human melanoma exosomes regulate the tumor immune response. Using microscopy and flow cytometry, we demonstrate that CD8+ T cells internalize exosomes from different tumor types even if these cells do not internalize vesicles as readily as other immune cells. We explored the function of melanoma-derived exosomes in CD8+ T cells and showed that these exosomes downregulate T-cell responses through decreased T-cell receptor (TCR) signaling and diminished cytokine and granzyme B secretions. The result reduces the cells' cytotoxic activity. Using mimics, we found that miRNAs enriched in exosomes-such as Homo sapiens (hsa)-miR-3187-3p, hsa-miR-498, hsa-miR-122, hsa-miR149, and hsa-miR-181a/b-regulate TCR signaling and TNFα secretion. Our observations suggest that miRNAs in melanoma-derived exosomes aid tumor immune evasion and could be a therapeutic target.
Collapse
Affiliation(s)
- Virginie Vignard
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France
| | - Maureen Labbé
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France
| | - Nadège Marec
- Plateforme CytoCell, SFR François Bonamy, Nantes, France
| | - Gwennan André-Grégoire
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Institut de Cancérologie de l'Ouest, Saint Herblain, France
| | - Nicolas Jouand
- Plateforme CytoCell, SFR François Bonamy, Nantes, France
| | - Jean-François Fonteneau
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France
| | - Nathalie Labarrière
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France
| | - Delphine Fradin
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France. .,LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France.,Cancéropole Grand-Ouest, Réseau Epigénétique (RepiCGO), Nantes, France
| |
Collapse
|
12
|
Courtney AH, Shvets AA, Lu W, Griffante G, Mollenauer M, Horkova V, Lo WL, Yu S, Stepanek O, Chakraborty AK, Weiss A. CD45 functions as a signaling gatekeeper in T cells. Sci Signal 2019; 12:12/604/eaaw8151. [PMID: 31641081 DOI: 10.1126/scisignal.aaw8151] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
T cells require the protein tyrosine phosphatase CD45 to detect and respond to antigen because it activates the Src family kinase Lck, which phosphorylates the T cell antigen receptor (TCR) complex. CD45 activates Lck by opposing the negative regulatory kinase Csk. Paradoxically, CD45 has also been implicated in suppressing TCR signaling by dephosphorylating the same signaling motifs within the TCR complex upon which Lck acts. We sought to reconcile these observations using chemical and genetic perturbations of the Csk/CD45 regulatory axis incorporated with computational analyses. Specifically, we titrated the activities of Csk and CD45 and assessed their influence on Lck activation, TCR-associated ζ-chain phosphorylation, and more downstream signaling events. Acute inhibition of Csk revealed that CD45 suppressed ζ-chain phosphorylation and was necessary for a regulatable pool of active Lck, thereby interconnecting the activating and suppressive roles of CD45 that tune antigen discrimination. CD45 suppressed signaling events that were antigen independent or induced by low-affinity antigen but not those initiated by high-affinity antigen. Together, our findings reveal that CD45 acts as a signaling "gatekeeper," enabling graded signaling outputs while filtering weak or spurious signaling events.
Collapse
Affiliation(s)
- Adam H Courtney
- Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA. .,Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alexey A Shvets
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wen Lu
- Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gloria Griffante
- Division of Molecular Immunology, Department of Internal Medicine, University Hospital Erlangen and Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | - Veronika Horkova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague 4, Czech Republic
| | - Wan-Lin Lo
- Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Steven Yu
- Howard Hughes Medical Institute (HHMI), San Francisco, CA 94143, USA
| | - Ondrej Stepanek
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague 4, Czech Republic
| | - Arup K Chakraborty
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA.,Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arthur Weiss
- Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA. .,Howard Hughes Medical Institute (HHMI), San Francisco, CA 94143, USA
| |
Collapse
|
13
|
Liang JW, Wang MY, Olounfeh KM, Zhao N, Wang S, Meng FH. Network pharmacology-based identifcation of potential targets of the flower of Trollius chinensis Bunge acting on anti-inflammatory effectss. Sci Rep 2019; 9:8109. [PMID: 31147584 PMCID: PMC6542797 DOI: 10.1038/s41598-019-44538-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 05/14/2019] [Indexed: 11/13/2022] Open
Abstract
The flower of Trollius chinensis Bunge was widely used for the treatment of inflammation-related diseases in traditional Chinese medicine (TCM). In order to clarify the anti-inflammatory mechanism of this Chinese herbs, a comprehensive network pharmacology strategy that consists of three sequential modules (pharmacophore matching, enrichment analysis and molecular docking.) was carried out. As a result, Apoptosis signal-regulating kinase 1 (ASK1), Janus kinase 1 (JAK1), c-Jun N-terminal kinases (JNKs), transforming protein p21 (HRas) and mitogen-activated protein kinase 14 (p38α) that related to the anti-inflammatory effect were filtered out. In further molecular dynamics (MD) simulation, the conformation of CID21578038 and CID20055288 were found stable in the protein ASK1 and JNKs respectively. The current investigation revealed that two effective compounds in the flower of Trollius chinensis Bunge played a crucial role in the process of inflammation by targeting ASK1 and JNKs, the comprehensive strategy can serve as a universal method to guide in illuminating the mechanism of the prescription of traditional Chinese medicine by identifying the pathways or targets.
Collapse
Affiliation(s)
- Jing-Wei Liang
- School of Pharmacy, China Medical University, Liaoning, 110122, China
| | - Ming-Yang Wang
- School of Pharmacy, China Medical University, Liaoning, 110122, China
| | | | - Nan Zhao
- School of Pharmacy, China Medical University, Liaoning, 110122, China
| | - Shan Wang
- School of Pharmacy, China Medical University, Liaoning, 110122, China
| | - Fan-Hao Meng
- School of Pharmacy, China Medical University, Liaoning, 110122, China.
| |
Collapse
|
14
|
Perron M, Saragovi HU. Inhibition of CD45 Phosphatase Activity Induces Cell Cycle Arrest and Apoptosis of CD45 + Lymphoid Tumors Ex Vivo and In Vivo. Mol Pharmacol 2018; 93:575-580. [PMID: 29555821 DOI: 10.1124/mol.117.110908] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 03/14/2018] [Indexed: 12/17/2022] Open
Abstract
Src-family kinases (SFK) govern cellular proliferation of bone marrow-derived cells. SFKs are regulated by the protein tyrosine phosphatase enzymatic activity of CD45. All lymphoid cells express CD45, but only proliferating cells are dependent on CD45 activity. We postulated that compound 211 (2-[(4-acetylphenyl)amino]-3-chloronaphthoquinone), a selective inhibitor of CD45 phosphatase activity, could preferentially affect actively proliferating cells but spare resting lymphoid cells. Compound 211 inhibited CD45 and induced inappropriate SFK signaling, leading to a G2/M cell cycle arrest and apoptotic cell death. CD45+ cell lines were sensitive to compound 211 cytotoxicity at low micromolar LD50 while control CD45- cell lines and CD45+ resting primary T cells were spared any toxicity. In two syngeneic tumor models in vivo, compound 211 delayed the growth of established primary tumors and reduced tumor metastasis without causing depletion of resting T cells. This work validates targeting CD45 phosphatase enzymatic activity, which may be a druggable target for cancer therapy.
Collapse
Affiliation(s)
- Michael Perron
- Lady Davis Institute-Jewish General Hospital and Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - H Uri Saragovi
- Lady Davis Institute-Jewish General Hospital and Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
15
|
CD45 in human physiology and clinical medicine. Immunol Lett 2018; 196:22-32. [PMID: 29366662 DOI: 10.1016/j.imlet.2018.01.009] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/18/2018] [Accepted: 01/19/2018] [Indexed: 01/20/2023]
Abstract
CD45 is an evolutionary highly conserved receptor protein tyrosine phosphatase exclusively expressed on all nucleated cells of the hematopoietic system. It is characterized by the expression of several isoforms, specific to a certain cell type and the developmental or activation status of the cell. CD45 is one of the key players in the initiation of T cell receptor signaling by controlling the activation of the Src family protein-tyrosine kinases Lck and Fyn. CD45 deficiency results in T- and B-lymphocyte dysfunction in the form of severe combined immune deficiency. It also plays a significant role in autoimmune diseases and cancer as well as in infectious diseases including fungal infections. The knowledge collected on CD45 biology is rather vast, but it remains unclear whether all findings in rodent immune cells also apply to human CD45. This review focuses on human CD45 expression and function and provides an overview on its ligands and role in human pathology.
Collapse
|
16
|
Yu Y, Sun H, Hou T, Wang S, Li Y. Fullerene derivatives act as inhibitors of leukocyte common antigen based on molecular dynamics simulations. RSC Adv 2018; 8:13997-14008. [PMID: 35539330 PMCID: PMC9079904 DOI: 10.1039/c7ra13543b] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/10/2018] [Indexed: 11/21/2022] Open
Abstract
Fullerene-based molecules are being studied as potential inhibitors of protein tyrosine phosphatases due to their unique properties and low toxicity. However, the underlying molecular mechanism remains elusive. In this study, molecular dynamics (MD) simulations in conjunction with molecular docking calculations were utilized to investigate the binding effects of C60, C60(NH2)30, and C60(OH)30 on the enzymatic activity of CD45 (a receptor-like protein tyrosine phosphatase). Our results show that all the investigated molecules can be docked into the region between D1 and D2 domains of CD45, and stabilize the protein structure. The average number of residues that directly interact with the C60(NH2)30 is two more than that of C60(OH)30, F819 and F820 (located in the loop connects α3 and β12), resulting in different effects of C60(NH2)30 and C60(OH)30 on protein activity. Detailed MD simulation analyses show that transformation of the interaction network caused by C60(NH2)30 is completely different from that of the control simulation due to the misfolding of α3. Furthermore, the movement of D1 active pocket and KNRY motif are most severely impaired by docking with C60(NH2)30. Our simulation results illustrate that fullerene derivatives modified with amino groups exhibit conspicuous tumor inhibition to protein tyrosine phosphatases, and can act as effective inhibitors. Our results give insight into the inhibitory effects of fullerene-based molecules on protein tyrosine phosphatases and providing a theoretical basis for the design of effective inhibitors. Fullerene-based molecules are being studied as potential inhibitors of protein tyrosine phosphatases due to their unique properties and low toxicity.![]()
Collapse
Affiliation(s)
- Yi Yu
- Institute of Functional Nano & Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- China
| | - Huiyong Sun
- Institute of Functional Nano & Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- China
| | - Tingjun Hou
- Institute of Functional Nano & Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- China
| | - Suidong Wang
- Institute of Functional Nano & Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- China
| | - Youyong Li
- Institute of Functional Nano & Soft Materials (FUNSOM)
- Soochow University
- Suzhou 215123
- China
| |
Collapse
|
17
|
Courtney AH, Lo WL, Weiss A. TCR Signaling: Mechanisms of Initiation and Propagation. Trends Biochem Sci 2017; 43:108-123. [PMID: 29269020 DOI: 10.1016/j.tibs.2017.11.008] [Citation(s) in RCA: 367] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/21/2017] [Accepted: 11/21/2017] [Indexed: 10/18/2022]
Abstract
The mechanisms by which a T cell detects antigen using its T cell antigen receptor (TCR) are crucial to our understanding of immunity and the harnessing of T cells therapeutically. A hallmark of the T cell response is the ability of T cells to quantitatively respond to antigenic ligands derived from pathogens while remaining inert to similar ligands derived from host tissues. Recent studies have revealed exciting properties of the TCR and the behaviors of its signaling effectors that are used to detect and discriminate between antigens. Here we highlight these recent findings, focusing on the proximal TCR signaling molecules Zap70, Lck, and LAT, to provide mechanistic models and insights into the exquisite sensitivity and specificity of the TCR.
Collapse
Affiliation(s)
- Adam H Courtney
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Howard Hughes Medical Institute (HHMI), San Francisco, CA 94143, USA
| | - Wan-Lin Lo
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Howard Hughes Medical Institute (HHMI), San Francisco, CA 94143, USA
| | - Arthur Weiss
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Howard Hughes Medical Institute (HHMI), San Francisco, CA 94143, USA.
| |
Collapse
|
18
|
Courtney AH, Amacher JF, Kadlecek TA, Mollenauer MN, Au-Yeung BB, Kuriyan J, Weiss A. A Phosphosite within the SH2 Domain of Lck Regulates Its Activation by CD45. Mol Cell 2017; 67:498-511.e6. [PMID: 28735895 DOI: 10.1016/j.molcel.2017.06.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 03/24/2017] [Accepted: 06/20/2017] [Indexed: 10/19/2022]
Abstract
The Src Family kinase Lck sets a critical threshold for T cell activation because it phosphorylates the TCR complex and the Zap70 kinase. How a T cell controls the abundance of active Lck molecules remains poorly understood. We have identified an unappreciated role for a phosphosite, Y192, within the Lck SH2 domain that profoundly affects the amount of active Lck in cells. Notably, mutation of Y192 blocks critical TCR-proximal signaling events and impairs thymocyte development in retrogenic mice. We determined that these defects are caused by hyperphosphorylation of the inhibitory C-terminal tail of Lck. Our findings reveal that modification of Y192 inhibits the ability of CD45 to associate with Lck in cells and dephosphorylate the C-terminal tail of Lck, which prevents its adoption of an active open conformation. These results suggest a negative feedback loop that responds to signaling events that tune active Lck amounts and TCR sensitivity.
Collapse
Affiliation(s)
- Adam H Courtney
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeanine F Amacher
- Departments of Molecular and Cell Biology and Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Theresa A Kadlecek
- The Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 04143, USA
| | - Marianne N Mollenauer
- The Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 04143, USA
| | - Byron B Au-Yeung
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - John Kuriyan
- Departments of Molecular and Cell Biology and Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA; The Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Arthur Weiss
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; The Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 04143, USA.
| |
Collapse
|
19
|
Loss of CD45 cell surface expression in canine T-zone lymphoma results from reduced gene expression. Vet Immunol Immunopathol 2017; 187:14-19. [DOI: 10.1016/j.vetimm.2017.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 02/28/2017] [Accepted: 03/23/2017] [Indexed: 12/19/2022]
|
20
|
Chang VT, Fernandes RA, Ganzinger KA, Lee SF, Siebold C, McColl J, Jönsson P, Palayret M, Harlos K, Coles CH, Jones EY, Lui Y, Huang E, Gilbert RJC, Klenerman D, Aricescu AR, Davis SJ. Initiation of T cell signaling by CD45 segregation at 'close contacts'. Nat Immunol 2016; 17:574-582. [PMID: 26998761 PMCID: PMC4839504 DOI: 10.1038/ni.3392] [Citation(s) in RCA: 217] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 12/29/2015] [Indexed: 12/14/2022]
Abstract
It has been proposed that the local segregation of kinases and the tyrosine phosphatase CD45 underpins T cell antigen receptor (TCR) triggering, but how such segregation occurs and whether it can initiate signaling is unclear. Using structural and biophysical analysis, we show that the extracellular region of CD45 is rigid and extends beyond the distance spanned by TCR-ligand complexes, implying that sites of TCR-ligand engagement would sterically exclude CD45. We also show that the formation of 'close contacts', new structures characterized by spontaneous CD45 and kinase segregation at the submicron-scale, initiates signaling even when TCR ligands are absent. Our work reveals the structural basis for, and the potent signaling effects of, local CD45 and kinase segregation. TCR ligands have the potential to heighten signaling simply by holding receptors in close contacts.
Collapse
Affiliation(s)
- Veronica T Chang
- Radcliffe Department of Medicine and MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Ricardo A Fernandes
- Radcliffe Department of Medicine and MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, United Kingdom
| | | | - Steven F Lee
- Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW
| | - Christian Siebold
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN
| | - James McColl
- Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW
| | - Peter Jönsson
- Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW
| | - Matthieu Palayret
- Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW
| | - Karl Harlos
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN
| | - Charlotte H Coles
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN
| | - E Yvonne Jones
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN
| | - Yuan Lui
- Radcliffe Department of Medicine and MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Elizabeth Huang
- Radcliffe Department of Medicine and MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Robert J C Gilbert
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW
| | - A Radu Aricescu
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN
| | - Simon J Davis
- Radcliffe Department of Medicine and MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, United Kingdom
| |
Collapse
|
21
|
Insights into the initiation of TCR signaling. Nat Immunol 2014; 15:798-807. [PMID: 25137454 DOI: 10.1038/ni.2940] [Citation(s) in RCA: 264] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 06/10/2014] [Indexed: 12/13/2022]
Abstract
The initiation of T cell antigen receptor signaling is a key step that can result in T cell activation and the orchestration of an adaptive immune response. Early events in T cell receptor signaling can distinguish between agonist and endogenous ligands with exquisite selectivity, and show extraordinary sensitivity to minute numbers of agonists in a sea of endogenous ligands. We review our current knowledge of models and crucial molecules that aim to provide a mechanistic explanation for these observations. Building on current understanding and a discussion of unresolved issues, we propose a molecular model for initiation of T cell receptor signaling that may serve as a useful guide for future studies.
Collapse
|
22
|
Furlan G, Minowa T, Hanagata N, Kataoka-Hamai C, Kaizuka Y. Phosphatase CD45 both positively and negatively regulates T cell receptor phosphorylation in reconstituted membrane protein clusters. J Biol Chem 2014; 289:28514-25. [PMID: 25128530 DOI: 10.1074/jbc.m114.574319] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
T cell receptor (TCR) phosphorylation requires the kinase Lck and phosphatase CD45. CD45 activates Lck by dephosphorylating an inhibitory tyrosine of Lck to relieve autoinhibition. However, CD45 also dephosphorylates the TCR, and the spatial exclusion of CD45 from TCR clustering in the plasma membrane appears to attenuate this negative effect of CD45. To further investigate the role of CD45 in signal initiation, we reconstituted membrane TCR clusters in vitro on supported lipid bilayers. Fluorescence microscopy of single clusters showed that incorporation of CD45 enhanced phosphorylation of TCR clusters, but only when Lck co-clustered with TCR. We found that clustered Lck autophosphorylated the inhibitory tyrosine and thus could be activated by CD45, whereas diffusive Lck molecules did not. In the TCR-Lck clusters and at low CD45 density, we speculate that the effect of Lck activation may overcome dephosphorylation of TCR, resulting in a net positive regulation. The CD45 density in physiological TCR clusters is also low because of the exclusion of CD45. Thus, we propose that the spatial organization of TCR/Lck/CD45 in T cell membranes is important not only for modulating the negative role of CD45 but also for creating conditions in which CD45 has a positive role in signal initiation.
Collapse
Affiliation(s)
- Gabriela Furlan
- From the International Center for Materials Nanoarchitectonics, National Institute for Materials Science, Tsukuba, Ibaraki 305-0047, Japan
| | - Takashi Minowa
- From the International Center for Materials Nanoarchitectonics, National Institute for Materials Science, Tsukuba, Ibaraki 305-0047, Japan
| | - Nobutaka Hanagata
- From the International Center for Materials Nanoarchitectonics, National Institute for Materials Science, Tsukuba, Ibaraki 305-0047, Japan
| | - Chiho Kataoka-Hamai
- From the International Center for Materials Nanoarchitectonics, National Institute for Materials Science, Tsukuba, Ibaraki 305-0047, Japan
| | - Yoshihisa Kaizuka
- From the International Center for Materials Nanoarchitectonics, National Institute for Materials Science, Tsukuba, Ibaraki 305-0047, Japan
| |
Collapse
|
23
|
Differentially-expressed genes identified by suppression subtractive hybridization in the bone marrow hematopoietic stem cells of patients with psoriasis. Mol Med Rep 2014; 10:479-85. [PMID: 24807678 DOI: 10.3892/mmr.2014.2203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Accepted: 02/24/2014] [Indexed: 11/05/2022] Open
Abstract
Psoriasis is a T cell-mediated, chronic, relapsing and inflammatory cutaneous disorder. The dysfunctional activity of T cells in patients with psoriasis is attributed to bone marrow hematopoietic stem cells (BMHSCs). To understand the pathogenic roles of BMHSCs in psoriasis, a differential gene expression analysis was performed using suppression subtractive hybridization of the BMHSCs from a patient with psoriasis and a healthy control. Using a cDNA array dot blot screening to screen 600 genes from forward- and reverse-subtracted cDNA libraries, 17 differentially-expressed sequence tags (ESTs) were identified. The genes within the ESTs were observed to be the homologs of genes that are involved in various cellular processes, including hormone signaling, RNA catabolism, protein ADP DNA base melting, transcriptional regulation, cell cycle regulation and metabolism. CD45, which was overexpressed in the psoriatic BMHSCs, was further analyzed using relative quantitative polymerase chain reaction. In addition, the levels of CD45 in the peripheral blood cells (PBCs) of the patients with psoriasis were markedly increased and closely associated with disease severity. An abnormality of hematopoietic progenitor cells, e.g., CD45 overexpression, may be transferred to PBCs via hematopoiesis, and may account for the psoriasis-inducing properties of activated T cells.
Collapse
|
24
|
Irving B, Weiss A. A Clue to Antigen Receptor Tails. THE JOURNAL OF IMMUNOLOGY 2014; 192:4013-4. [DOI: 10.4049/jimmunol.1400660] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
Perron MD, Chowdhury S, Aubry I, Purisima E, Tremblay ML, Saragovi HU. Allosteric noncompetitive small molecule selective inhibitors of CD45 tyrosine phosphatase suppress T-cell receptor signals and inflammation in vivo. Mol Pharmacol 2014; 85:553-63. [PMID: 24473749 DOI: 10.1124/mol.113.089847] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
CD45 is a receptor-like member of the protein tyrosine phosphatase (PTP) family. We screened in silico for small molecules binding at a predicted allosteric pocket unique to the CD45 intracellular domain, and validated inhibitors by in vitro phosphatase assays. Compound 211 exhibited a CD45 IC50 value of 200 nM and had >100-fold selectivity over six related PTPs. The relevance of the allosteric pocket was verified through site-directed mutagenesis. Compound 211 has a noncompetitive mechanism of action, and it is extremely effective at preventing dephosphorylation of substrate Lck phosphotyrosine (pY)-505 versus preventing dephosphorylation of Lck pY-393. In cultured primary T cells, compound 211 prevents T-cell receptor-mediated activation of Lck, Zap-70, and mitogen-activated protein kinase, and interleukin-2 production. In a delayed-type hypersensitivity reaction in vivo, compound 211 abolished inflammation. This work demonstrates a novel approach to develop effective allosteric inhibitors that can be expanded to target the corresponding allosteric domains of other receptor PTPs.
Collapse
Affiliation(s)
- Michael D Perron
- Lady Davis Institute-Jewish General Hospital (M.P., S.C., H.U.S.), Departments of Pharmacology and Therapeutics (M.P., H.U.S.), Biochemistry (I.A., E.P., M.L.T.), and Oncology (H.U.S.), Goodman Cancer Research Center (M.L.T.), and Segal Cancer Center, McGill University, Montreal, Quebec, Canada (H.U.S.); and Biotechnology Research Institute, National Research Council Canada, Montreal, Quebec, Canada (E.P.)
| | | | | | | | | | | |
Collapse
|
26
|
Kouroupis D, Baboolal TG, Jones E, Giannoudis PV. Native multipotential stromal cell colonization and graft expander potential of a bovine natural bone scaffold. J Orthop Res 2013; 31:1950-8. [PMID: 23868185 DOI: 10.1002/jor.22438] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 06/24/2013] [Indexed: 02/04/2023]
Abstract
Graft expanders are bone scaffolds used, in combination with autografts, to fill large bone defects in trauma surgery. This study investigates the graft expander potential of a natural bone substitute Orthoss by studying its ability to support attachment, growth and osteogenic differentiation of neighboring multipotential stromal cells (MSCs). Material consisting of bone marrow (BM) aspirate and reamer-irrigator-aspirator (RIA)-harvested autograft bone was co-cultured with commercially available Orthoss granules. Native MSCs attached to Orthoss were expanded and phenotypically characterized. MSCs egress from neighboring cancelous bone was assessed in 3D Matrigel co-cultures. MSC differentiation was evaluated using scanning electron microscopy and measuring alkaline phosphatase (ALP) activity per cell. CD45(+) hematopoietic lineage cells and highly proliferative CD90(+) CD73(+) CD105(+) MSCs preferentially colonized Orthoss granules, over RIA bone chips. MSC colonization was followed by their intrinsic osteogenic differentiation, assessed as mineral deposition and gradual rise in ALP activity, even in the absence of osteogenic stimuli. When in contact with mixed cell populations and RIA chips, Orthoss granules support the attachment, growth and osteogenic differentiation of neighboring MSCs. Therefore, natural bone substitutes similar to Orthoss can be used as void fillers and graft expanders for repairing large bone defects in conjunction with autologous BM aspirates and autografts.
Collapse
Affiliation(s)
- Dimitrios Kouroupis
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | | | | | | |
Collapse
|
27
|
Kharitidi D, Manteghi S, Pause A. Pseudophosphatases: methods of analysis and physiological functions. Methods 2013; 65:207-18. [PMID: 24064037 DOI: 10.1016/j.ymeth.2013.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 08/20/2013] [Accepted: 09/11/2013] [Indexed: 01/27/2023] Open
Abstract
Protein tyrosine phosphatases (PTPs) are key enzymes in the regulation of cellular homeostasis and signaling pathways. Strikingly, not all PTPs bear enzymatic activity. A considerable fraction of PTPs are enzymatically inactive and are known as pseudophosphatases. Despite the lack of activity they execute pivotal roles in development, cell biology and human disease. The present review is focused on the methods used to identify pseudophosphatases, their targets, and physiological roles. We present a strategy for detailed enzymatic analysis of inactive PTPs, regulation of inactive PTP domains and identification of binding partners. Furthermore, we provide a detailed overview of human pseudophosphatases and discuss their regulation of cellular processes and functions in human pathologies.
Collapse
Affiliation(s)
- Dmitri Kharitidi
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, 3655, Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada.
| | - Sanaz Manteghi
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, 3655, Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada.
| | - Arnim Pause
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, 3655, Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
28
|
Heterologous immunity triggered by a single, latent virus in Mus musculus: combined costimulation- and adhesion- blockade decrease rejection. PLoS One 2013; 8:e71221. [PMID: 23940724 PMCID: PMC3733932 DOI: 10.1371/journal.pone.0071221] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 06/26/2013] [Indexed: 01/11/2023] Open
Abstract
The mechanisms underlying latent-virus-mediated heterologous immunity, and subsequent transplant rejection, especially in the setting of T cell costimulation blockade, remain undetermined. To address this, we have utilized MHV68 to develop a rodent model of latent virus-induced heterologous alloimmunity. MHV68 infection was correlated with multimodal immune deviation, which included increased secretion of CXCL9 and CXCL10, and with the expansion of a CD8(dim) T cell population. CD8(dim) T cells exhibited decreased expression of multiple costimulation molecules and increased expression of two adhesion molecules, LFA-1 and VLA-4. In the setting of MHV68 latency, recipients demonstrated accelerated costimulation blockade-resistant rejection of skin allografts compared to non-infected animals (MST 13.5 d in infected animals vs 22 d in non-infected animals, p<.0001). In contrast, the duration of graft acceptance was equivalent between non-infected and infected animals when treated with combined anti-LFA-1/anti-VLA-4 adhesion blockade (MST 24 d for non-infected and 27 d for infected, p = n.s.). The combination of CTLA-4-Ig/anti-CD154-based costimulation blockade+anti-LFA-1/anti-VLA-4-based adhesion blockade led to prolonged graft acceptance in both non-infected and infected cohorts (MST>100 d for both, p<.0001 versus costimulation blockade for either). While in the non-infected cohort, either CTLA-4-Ig or anti-CD154 alone could effectively pair with adhesion blockade to prolong allograft acceptance, in infected animals, the prolonged acceptance of skin grafts could only be recapitulated when anti-LFA-1 and anti-VLA-4 antibodies were combined with anti-CD154 (without CTLA-4-Ig, MST>100 d). Graft acceptance was significantly impaired when CTLA-4-Ig alone (no anti-CD154) was combined with adhesion blockade (MST 41 d). These results suggest that in the setting of MHV68 infection, synergy occurs predominantly between adhesion pathways and CD154-based costimulation, and that combined targeting of both pathways may be required to overcome the increased risk of rejection that occurs in the setting of latent-virus-mediated immune deviation.
Collapse
|
29
|
Tonks NK. Protein tyrosine phosphatases--from housekeeping enzymes to master regulators of signal transduction. FEBS J 2013; 280:346-78. [PMID: 23176256 DOI: 10.1111/febs.12077] [Citation(s) in RCA: 354] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 11/13/2012] [Accepted: 11/15/2012] [Indexed: 12/12/2022]
Abstract
There are many misconceptions surrounding the roles of protein phosphatases in the regulation of signal transduction, perhaps the most damaging of which is the erroneous view that these enzymes exert their effects merely as constitutively active housekeeping enzymes. On the contrary, the phosphatases are critical, specific regulators of signalling in their own right and serve an essential function, in a coordinated manner with the kinases, to determine the response to a physiological stimulus. This review is a personal perspective on the development of our understanding of the protein tyrosine phosphatase family of enzymes. I have discussed various aspects of the structure, regulation and function of the protein tyrosine phosphatase family, which I hope will illustrate the fundamental importance of these enzymes in the control of signal transduction.
Collapse
Affiliation(s)
- Nicholas K Tonks
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724-2208, USA.
| |
Collapse
|
30
|
He R, Zeng LF, He Y, Zhang S, Zhang ZY. Small molecule tools for functional interrogation of protein tyrosine phosphatases. FEBS J 2012; 280:731-50. [PMID: 22816879 DOI: 10.1111/j.1742-4658.2012.08718.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The importance of protein tyrosine phosphatases (PTPs) in the regulation of cellular signalling is well established. Malfunction of PTP activity is also known to be associated with cancer, metabolic syndromes and autoimmune disorders, as well as neurodegenerative and infectious diseases. However, a detailed understanding of the roles played by the PTPs in normal physiology and in pathogenic conditions has been hampered by the absence of PTP-specific small molecule agents. In addition, the therapeutic benefits of modulating this target class are underexplored as a result of a lack of suitable chemical probes. Potent and specific PTP inhibitors could significantly facilitate functional analysis of the PTPs in complex cellular signal transduction pathways and may constitute valuable therapeutics in the treatment of several human diseases. We highlight the current challenges to and opportunities for developing PTP-specific small molecule agents. We also review available selective small molecule inhibitors developed for a number of PTPs, including PTP1B, TC-PTP, SHP2, lymphoid-specific tyrosine phosphatase, haematopoietic protein tyrosine phosphatase, CD45, PTPβ, PTPγ, PTPRO, Vaccinia H1-related phosphatase, mitogen-activated protein kinase phosphatase-1, mitogen-activated protein kinase phosphatase-3, Cdc25, YopH, mPTPA and mPTPB.
Collapse
Affiliation(s)
- Rongjun He
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | |
Collapse
|
31
|
Wiede F, Shields BJ, Chew SH, Kyparissoudis K, van Vliet C, Galic S, Tremblay ML, Russell SM, Godfrey DI, Tiganis T. T cell protein tyrosine phosphatase attenuates T cell signaling to maintain tolerance in mice. J Clin Invest 2011; 121:4758-74. [PMID: 22080863 PMCID: PMC3226006 DOI: 10.1172/jci59492] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 10/07/2011] [Indexed: 12/14/2022] Open
Abstract
Many autoimmune diseases exhibit familial aggregation, indicating that they have genetic determinants. Single nucleotide polymorphisms in PTPN2, which encodes T cell protein tyrosine phosphatase (TCPTP), have been linked with the development of several autoimmune diseases, including type 1 diabetes and Crohn's disease. In this study, we have identified TCPTP as a key negative regulator of TCR signaling, which might explain the association of PTPN2 SNPs with autoimmune disease. We found that TCPTP dephosphorylates and inactivates Src family kinases to regulate T cell responses. Using T cell-specific TCPTP-deficient mice, we established that TCPTP attenuates T cell activation and proliferation in vitro and blunts antigen-induced responses in vivo. TCPTP deficiency lowered the in vivo threshold for TCR-dependent CD8(+) T cell proliferation. Consistent with this, T cell-specific TCPTP-deficient mice developed widespread inflammation and autoimmunity that was transferable to wild-type recipient mice by CD8(+) T cells alone. This autoimmunity was associated with increased serum levels of proinflammatory cytokines and anti-nuclear antibodies, T cell infiltrates in non-lymphoid tissues, and liver disease. These data indicate that TCPTP is a critical negative regulator of TCR signaling that sets the threshold for TCR-induced naive T cell responses to prevent autoimmune and inflammatory disorders arising.
Collapse
MESH Headings
- Animals
- Antibodies, Antinuclear/biosynthesis
- Autoimmune Diseases/enzymology
- Autoimmune Diseases/etiology
- Autoimmune Diseases/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/transplantation
- Immune Tolerance/immunology
- Inflammation/blood
- Inflammation/genetics
- Inflammation/immunology
- Lymphocyte Activation
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Phosphorylation
- Protein Processing, Post-Translational
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/deficiency
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology
- Radiation Chimera
- Receptors, Antigen, T-Cell/immunology
- Signal Transduction/immunology
- T-Lymphocyte Subsets/enzymology
- T-Lymphocyte Subsets/immunology
- Thymocytes/pathology
- ZAP-70 Protein-Tyrosine Kinase/physiology
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Florian Wiede
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Benjamin J. Shields
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Sock Hui Chew
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Konstantinos Kyparissoudis
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Catherine van Vliet
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Sandra Galic
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Michel L. Tremblay
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Sarah M. Russell
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Dale I. Godfrey
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Tony Tiganis
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia.
McGill Cancer Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
Immune Signaling Laboratory, Cancer Immunology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
Centre for MicroPhotonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
| |
Collapse
|
32
|
Maisey K, Imarai M. Diversity of teleost leukocyte molecules: role of alternative splicing. FISH & SHELLFISH IMMUNOLOGY 2011; 31:663-672. [PMID: 20723604 DOI: 10.1016/j.fsi.2010.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 08/05/2010] [Accepted: 08/09/2010] [Indexed: 05/29/2023]
Abstract
Alternative splicing is an important mechanism of gene expression control that also produces a large proteome from a limited number of genes. In the immune system of mammals, numerous relevant genes have been found to undergo alternative splicing that contributes to the complexity of immune response. An increasing number of reports have recently indicated that alternative splicing also occurs in other vertebrates, such as fish. In this review we summarize the general features of such molecular events in cytokines and leukocyte co-receptors and their contribution to diversity and regulation of fish leukocytes.
Collapse
Affiliation(s)
- Kevin Maisey
- Laboratorio de Inmunología, Centro de Biotecnología Acuícola (CBA), Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Correo 40, Casilla 33, Santiago, Chile.
| | | |
Collapse
|
33
|
Herpes simplex virus requires VP11/12 to activate Src family kinase-phosphoinositide 3-kinase-Akt signaling. J Virol 2011; 85:2803-12. [PMID: 21228233 DOI: 10.1128/jvi.01877-10] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
We previously showed that the herpes simplex virus 1 (HSV-1) tegument protein VP11/12 activates the lymphocyte-specific Src family kinase (SFK) Lck and is tyrosine phosphorylated in an Lck-dependent manner during T cell infection. We now extend these findings to show that ectopic expression of Lck induces robust tyrosine phosphorylation of VP11/12 in Vero cells, strongly suggesting that VP11/12 participates in an Lck-mediated signaling pathway as a substrate of Lck or a kinase activated by Lck. We sought to elucidate signaling events downstream of VP11/12-SFK interactions. SFKs lie upstream of the canonical phosphoinositide 3-kinase (PI3K)-Akt pathway in signaling emanating from immune receptors, growth factor receptors, and polyomavirus middle T antigen. Here, we show that VP11/12 is required for virus-induced activation of PI3K-Akt signaling in HSV-infected Jurkat T cells and primary fibroblasts. VP11/12 interacts with PI3K or PI3K signaling complexes during infection, suggesting that VP11/12 activates PI3K directly. SFK activity is required for tyrosine phosphorylation of VP11/12, VP11/12-PI3K interactions, and Akt activation in infected fibroblasts, suggesting that SFK-dependent phosphorylation of VP11/12 is required for interactions with downstream signaling effectors. Akt controls many biological functions, including cell survival, cell motility, and translation, but it is currently unclear which Akt targets are modulated by VP11/12 during infection. Although the Akt target mTORC1 is activated during HSV-1 infection, VP11/12 is not required for this effect, implying that one or more additional viral proteins regulate this pathway. Further studies are therefore required to determine which Akt targets and associated biological functions are uniquely modulated by VP11/12.
Collapse
|
34
|
Dupéré-Minier G, Desharnais P, Bernier J. Involvement of tyrosine phosphatase CD45 in apoptosis. Apoptosis 2010; 15:1-13. [PMID: 19856105 DOI: 10.1007/s10495-009-0413-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CD45 is a transmembrane molecule with phosphatase activity expressed in all nucleated haematopoietic cells and plays a major role in immune cells. It is a protein tyrosine phosphatase that is essential for antigen-receptor-mediated signal transduction by regulating Src family members that initiate TCR signaling. CD45 is being attributed a new emerging role as an apoptosis regulator. Cross-linking of the extracellular portion of the CD45 by monoclonal antibodies and by galectin-1, can induce apoptosis in T and B cells. Interestingly, this phosphatase has also been involved in nuclear apoptosis induced by mitochondrial perturbing agents. Furthermore, it is involved in apoptosis induced by HIV-1. CD45 defect is implicated in various diseases such as severe-combined immunodeficiency disease (SCID), acquired immunodeficiency syndrome (AIDS), lymphoma and multiple myelomas. The understanding of the mechanisms by which CD45 regulates apoptosis would be very useful in disease treatment.
Collapse
|
35
|
Saunders AE, Johnson P. Modulation of immune cell signalling by the leukocyte common tyrosine phosphatase, CD45. Cell Signal 2010; 22:339-48. [PMID: 19861160 DOI: 10.1016/j.cellsig.2009.10.003] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 10/18/2009] [Indexed: 01/01/2023]
Abstract
CD45 is a leukocyte specific transmembrane glycoprotein and a receptor-like protein tyrosine phosphatase (PTP). CD45 can be expressed as several alternatively spliced isoforms that differ in the extracellular domain. The isoforms are regulated in a cell type and activation state-dependent manner, yet their function has remained elusive. The Src family kinase members Lck and Lyn are key substrates for CD45 in T and B lymphocytes, respectively. CD45 lowers the threshold of antigen receptor signalling, which impacts T and B cell activation and development. CD45 also regulates antigen triggered Fc receptor signalling in mast cells and Toll-like receptor (TLR) signalling in dendritic cells, thus broadening the role of CD45 to other recognition receptors involved in adaptive and innate immunity. In addition, CD45 can affect immune cell adhesion and migration and can modulate cytokine production and signalling. Here we review what is known about the substrate specificity and regulation of CD45 and summarise its effect on immune cell signalling pathways, from its established role in T and B antigen receptor signalling to its emerging role regulating innate immune cell recognition and cytokine production.
Collapse
Affiliation(s)
- A E Saunders
- Department of Microbiology and Immunology, Life Sciences Institute, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada V6T 1Z3
| | | |
Collapse
|
36
|
The right team at the right time to go for a home run: tyrosine kinase activation by the TCR. Nat Immunol 2010; 11:101-4. [PMID: 20084063 DOI: 10.1038/ni0210-101] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The TCR signals via cytoplasmic tyrosine kinases. Art Weiss recounts discoveries that led to early understanding of these events.
Collapse
|
37
|
Seki I, Suzuki M, Miyasaka N, Kohsaka H. Expression of CD45 isoforms correlates with differential proliferative responses of peripheral CD4+ and CD8+ T cells. Immunol Lett 2010; 129:39-46. [PMID: 20093141 DOI: 10.1016/j.imlet.2009.12.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 12/02/2009] [Accepted: 12/23/2009] [Indexed: 11/28/2022]
Abstract
CD4(+) T cells express IL-2 receptor complexes to the same level as CD8(+) T cells when the two T cell populations were stimulated simultaneously. However, the activation of downstream signaling molecules, such as Jaks, was increased in CD8(+) T cells. Although equivalent amounts of CD45, which acts as a Jak phosphatase, was expressed on the two T cell populations, those on the CD8(+) T cells have less protein tyrosine phosphatase activity than those on the CD4(+) T cells. Furthermore, we find that different CD45 isoforms dominate in the two populations; CD45RO on proliferating CD4(+) T cells and CD45RBC on proliferating CD8(+) T cells. In addition, NIH3T3 cells expressing the CD45RBC transgene had more phosphorylated Jak1 and grew faster than those with the CD45RO transgene. Thus, the expression of specific CD45 isoforms on T cells correlates with their proliferative response to IL-2, suggesting that controlling cells expressing specific CD45 isoforms could correct excessive or insufficient immune responses.
Collapse
Affiliation(s)
- Iwao Seki
- Department of Medicine and Rheumatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | |
Collapse
|
38
|
Falahati R, Leitenberg D. Selective regulation of TCR signaling pathways by the CD45 protein tyrosine phosphatase during thymocyte development. THE JOURNAL OF IMMUNOLOGY 2009; 181:6082-91. [PMID: 18941197 DOI: 10.4049/jimmunol.181.9.6082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In CD45-deficient animals, there is a severe defect in thymocyte-positive selection, resulting in an absence of mature T cells and the accumulation of thymocytes at the DP stage of development. However, the signaling defect(s) responsible for the block in development of mature single-positive T cells is not well characterized. Previous studies have found that early signal transduction events in CD45-deficient cell lines and thymocytes are markedly diminished following stimulation with anti-CD3. Nevertheless, there are also situations in which T cell activation and TCR signaling events can be induced in the absence of CD45. For example, CD45-independent TCR signaling can be recovered upon simultaneous Ab cross-linking of CD3 and CD4 compared with cross-linking of CD3 alone. These data suggest that CD45 may differentially regulate TCR signaling events depending on the nature of the signal and/or on the differentiation state of the cell. In the current study, we have assessed the role of CD45 in regulating primary thymocyte activation following physiologic stimulation with peptide. Unlike CD3-mediated stimulation, peptide stimulation of CD45-deficient thymocytes induces diminished, but readily detectable TCR-mediated signaling events, such as phosphorylation of TCR-associated zeta, ZAP70, linker for activation of T cells, and Akt, and increased intracellular calcium concentration. In contrast, phosphorylation of ERK, which is essential for positive selection, is more severely affected in the absence of CD45. These data suggest that CD45 has a selective role in regulating different aspects of T cell activation.
Collapse
Affiliation(s)
- Rustom Falahati
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC 20037, USA
| | | |
Collapse
|
39
|
Spinozzi F, Agea E, Fiorucci G, Gerli R, Muscat C, Belia S, Bertotto A. Ethanol-induced CD3 and CD2 hyporesponsiveness of peripheral blood T lymphocytes. Immunopharmacol Immunotoxicol 2008; 14:939-53. [PMID: 1363475 DOI: 10.3109/08923979209009243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The functional relevance of a direct ethanol effect on the membrane structure of T lymphocytes and accessory cells (APC), as well as on signal transduction systems was studied in ten normal subjects. Ethanol incubation (80 mM for 24h) of highly purified T cells increased the number of CD4+/CD45RA+ lymphocytes. In contrast, ethanol exposure induced a drop in CD14+/LFA-3+ APC values. These changes were accompanied by faulty T-cell proliferation in response to anti-CD3 and anti-CD2 mAb and inhibition of CD3- and CD2-mediated rises in intracellular calcium and, to a lesser extent, inositol 1,4,5-triphosphate levels. These data clearly indicate that a membrane-specific ethanol interaction both modifies surface glycoproteic and/or glycolipidic structures and alters transmembrane transduction of the activation signals.
Collapse
Affiliation(s)
- F Spinozzi
- Department of Internal Medicine, University of Perugia, Italy
| | | | | | | | | | | | | |
Collapse
|
40
|
The cytoplasmic tail of CD45 is released from activated phagocytes and can act as an inhibitory messenger for T cells. Blood 2008; 112:1240-8. [DOI: 10.1182/blood-2008-02-138131] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AbstractCD45 is the prototypic transmembrane protein tyrosine phosphatase (PTP), which is expressed on all nucleated hematopoietic cells and plays a central role in the integration of environmental signals into immune cell responses. Here we report an alternative function for the intracellular domain of CD45. We dis-covered that CD45 is sequentially cleaved by serine/metalloproteinases and γ-secretases during activation of human monocytes and granulocytes by fungal stimuli or phorbol 12-myristate 13-acetate but not by other microbial stimuli. Proteolytic processing of CD45 occurred upon activation of monocytes or granulocytes but not of T cells, B cells, or dendritic cells and resulted in a 95-kDa fragment of the cytoplasmic tail of CD45 (ct-CD45). ct-CD45 was released from monocytes and granulocytes upon activation-induced cell death. Binding studies with ct-CD45 revealed a counter-receptor on preactivated T cells. Moreover, T-cell proliferation induced by dendritic cells or CD3 antibodies was inhibited in the presence of ct-CD45. Taken together, the results of our study demonstrate that fragments of the intracellular domain of CD45 from human phagocytes can function as intercellular regulators of T-cell activation.
Collapse
|
41
|
O'Shea JJ, Johnston JA, Kehrl J, Koretzky G, Samelson LE. Key molecules involved in receptor-mediated lymphocyte activation. ACTA ACUST UNITED AC 2008; Chapter 11:Unit 11.9A. [PMID: 18432708 DOI: 10.1002/0471142735.im1109as44] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
This unit, along with Unit 11.9B, provides a summary of our current knowledge about various signaling pathways critical to the function of immune cells. Here, our understanding of T cell receptor (TCR)- and B cell receptor (BCR)-mediated signaling is summarized. A schematic representation of immunologically relevant cytokine receptors and the Janus Family Kinases (JAKs) that is activated through these receptors is provided, along with details about molecules involved in interleukin 2 mediated signal transduction.
Collapse
Affiliation(s)
- J J O'Shea
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | | | | | | | | |
Collapse
|
42
|
Nika K, Tautz L, Arimura Y, Vang T, Williams S, Mustelin T. A weak Lck tail bite is necessary for Lck function in T cell antigen receptor signaling. J Biol Chem 2007; 282:36000-9. [PMID: 17897955 DOI: 10.1074/jbc.m702779200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Src family kinases are suppressed by a "tail bite" mechanism, in which the binding of a phosphorylated tyrosine in the C terminus of the protein to the Src homology (SH) 2 domain in the N-terminal half of the protein forces the catalytic domain into an inactive conformation stabilized by an additional SH3 interaction. In addition to this intramolecular suppressive function, the SH2 domain also mediates intermolecular interactions, which are crucial for T cell antigen receptor (TCR) signaling. To better understand the relative importance of these two opposite functions of the SH2 domain of the Src family kinase Lck in TCR signaling, we created three mutants of Lck in which the intramolecular binding of the C terminus to the SH2 domain was strengthened. The mutants differed from wild-type Lck only in one to three amino acid residues following the negative regulatory tyrosine 505, which was normally phosphorylated by Csk and dephosphorylated by CD45 in the mutants. In the Lck-negative JCaM1 cell line, the Lck mutants had a much reduced ability to transduce signals from the TCR in a manner that directly correlated with SH2-Tyr(P)(505) affinity. The mutant with the strongest tail bite was completely unable to support any ZAP-70 phosphorylation, mitogen-activated protein kinase activation, or downstream gene activation in response to TCR ligation, whereas other mutants had intermediate abilities. Lipid raft targeting was not affected. We conclude that Lck is regulated by a weak tail bite to allow for its activation and service in TCR signaling, perhaps through a competitive SH2 engagement mechanism.
Collapse
Affiliation(s)
- Konstantina Nika
- Program on Inflammatory Disease Research, Infectious and Inflammatory Disease Center, The Burnham Institute for Medical Research, La Jolla, California 92037, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Grande SM, Bannish G, Fuentes-Panana EM, Katz E, Monroe JG. Tonic B-cell and viral ITAM signaling: context is everything. Immunol Rev 2007; 218:214-34. [PMID: 17624955 DOI: 10.1111/j.1600-065x.2007.00535.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The presence of an immunoreceptor tyrosine-based activation motif (ITAM) makes immunoreceptors different from other signaling receptors, like integrins, G-coupled protein receptors, chemokine receptors, and growth factor receptors. This unique motif has the canonical sequence D/Ex(0-2)YxxL/Ix(6-8)YxxL/I, where x represents any amino acid and is present at least once in all immunoreceptor complexes. Immunoreceptors can promote survival, activation, and differentiation by transducing signals through these highly conserved motifs. Traditionally, ITAM signaling is thought to occur in response to ligand-induced aggregation, although evidence indicates that ligand-independent tonic signaling also provides functionally relevant signals. The majority of proteins containing ITAMs are transmembrane proteins that exist as part of immunoreceptor complexes. However, oncogenic viruses also have ITAM-containing proteins. In this review, we discuss what is known about tonic signaling by both cellular and viral ITAM-containing proteins and speculate what we might learn from each context.
Collapse
Affiliation(s)
- Shannon M Grande
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
44
|
Abstract
The discovery that a single amino acid substitution in the PTPN22 protein tyrosine phosphatase can predispose to so many autoimmune diseases (see chapters 2 and 3), even when present in a single copy, raises many questions regarding the broader significance of this observation. Is there something unique about PTPN22 or are genetic variants of other protein tyrosine phosphatases likely also associated with autoimmune disease? If so, will polymorphisms in other phosphatases be found in the same spectrum of diseases? Are protein tyrosine phosphatases like PTPN22 good drug targets for the treatment of human autoimmunity? In this review, I offer some basis for thinking about these questions.
Collapse
Affiliation(s)
- Tomas Mustelin
- Program on Inflammatory Disease Research Infectious and Inflammatory Disease Center, The Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
45
|
Hermiston ML, Tan AL, Gupta VA, Majeti R, Weiss A. The juxtamembrane wedge negatively regulates CD45 function in B cells. Immunity 2006; 23:635-47. [PMID: 16356861 DOI: 10.1016/j.immuni.2005.11.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2005] [Revised: 09/08/2005] [Accepted: 11/09/2005] [Indexed: 12/22/2022]
Abstract
CD45 is a receptor-like protein tyrosine phosphatase highly expressed on all nucleated hematopoietic cells. We previously generated mice containing a point mutation in the juxtamembrane wedge of CD45. Demonstrating the critical negative regulatory function of the wedge, the CD45 E613R mutation led to a lymphoproliferative disorder (LPD) and a lupus-like autoimmune syndrome. Here we show the central role of B cells in this phenotype. Genetic elimination of B cells, but not T cells, ablates the LPD. In contrast to CD45-deficient B cells, the E613R mutation generates hyperresponsive B cells. Comparison of CD45-deficient and CD45 E613R mice reveals dichotomous effects of these mutations on B cell development. Together, the results support a role for CD45 as a rheostat, with both positive and negative regulatory functions, that fine-tunes the signal transduction threshold at multiple checkpoints in B cell development.
Collapse
Affiliation(s)
- Michelle L Hermiston
- Department of Pediatrics, University of California, San Francisco, San Francisco, California 94143, USA
| | | | | | | | | |
Collapse
|
46
|
Deng S, Moore DJ, Huang X, Mohiuddin M, Lee MK, Velidedeoglu E, Lian MM, Chiaccio M, Sonawane S, Orlin A, Wang J, Chen H, Caton A, Zhong R, Markmann JF. Antibody-induced transplantation tolerance that is dependent on thymus-derived regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2006; 176:2799-807. [PMID: 16493036 DOI: 10.4049/jimmunol.176.5.2799] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Targeting of the CD45RB isoform by mAb (anti-CD45RB) effectively induces donor-specific tolerance to allografts. The immunological mechanisms underlying the tolerant state remain unclear although some studies have suggested the involvement of regulatory T cells (T-regs). Although their generative pathway remains undefined, tolerance promoting T-regs induced by systemic anti-CD45RB treatment have been assumed to originate in the peripheral immune system. We demonstrate herein that separable effects on the peripheral and central immune compartments mediate graft survival induced by anti-CD45RB administration. In the absence of the thymus, anti-CD45RB therapy is not tolerogenic though it retains peripheral immunosuppressive activity. The thymus is required for anti-CD45RB to produce indefinite graft survival and donor-specific tolerance, and this effect is accomplished through thymic production of donor-specific T-regs. These data reveal for the first time an Ab-based tolerance regimen that relies on the central tolerance pathway.
Collapse
Affiliation(s)
- Shaoping Deng
- Harrison Department of Surgical Research, Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Maksumova L, Le HT, Muratkhodjaev F, Davidson D, Veillette A, Pallen CJ. Protein Tyrosine Phosphatase α Regulates Fyn Activity and Cbp/PAG Phosphorylation in Thymocyte Lipid Rafts. THE JOURNAL OF IMMUNOLOGY 2005; 175:7947-56. [PMID: 16339530 DOI: 10.4049/jimmunol.175.12.7947] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A role for the receptor protein tyrosine phosphatase alpha (PTPalpha) in immune cell function and regulation of Src family kinases was investigated using thymocytes from PTPalpha-deficient mice. PTPalpha-null thymocytes develop normally, but unstimulated PTPalpha-/- cells exhibit increased tyrosine phosphorylation of specific proteins, increased Fyn activity, and hyperphosphorylation of Cbp/PAG that promotes its association with C-terminal Src kinase. Elevated Fyn activity in the absence of PTPalpha is due to enhanced phosphorylation of Fyn tyrosines 528 and 417. Some PTPalpha is localized in lipid rafts of thymocytes, and raft-associated Fyn is specifically activated in PTPalpha-/- cells. PTPalpha is not a Cbp/PAG phosphatase, because it is not required for Cbp/PAG dephosphorylation in unstimulated or anti-CD3-stimulated thymocytes. Together, our results indicate that PTPalpha, likely located in lipid rafts, regulates the activity of raft Fyn. In the absence of PTPalpha this population of Fyn is activated and phosphorylates Cbp/PAG to enhance association with C-terminal Src kinase. Although TCR-mediated tyrosine phosphorylation was apparently unaffected by the absence of PTPalpha, the long-term proliferative response of PTPalpha-/- thymocytes was reduced. These findings indicate that PTPalpha is a component of the complex Src family tyrosine kinase regulatory network in thymocytes and is required to suppress Fyn activity in unstimulated cells in a manner that is not compensated for by the major T cell PTP and SFK regulator, CD45.
Collapse
Affiliation(s)
- Lola Maksumova
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Zhang ZY. Functional studies of protein tyrosine phosphatases with chemical approaches. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2005; 1754:100-7. [PMID: 16226063 DOI: 10.1016/j.bbapap.2005.09.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2005] [Revised: 09/05/2005] [Accepted: 09/05/2005] [Indexed: 01/24/2023]
Abstract
Protein tyrosine phosphatases (PTPs) are important signaling enzymes that serve as key regulatory components in signal transduction pathways. Defective or inappropriate regulation of PTP activity leads to aberrant tyrosine phosphorylation, which contributes to the development of many human diseases. A number of PTPs have been identified as novel therapeutic targets for the treatment of various diseases. However, because PTPs can both enhance and antagonize PTK signaling, it is essential to elucidate the physiological context in which PTPs function. Assigning the functional significance of PTPs in normal physiology and in diseases remains a major challenge in cell signaling. Efficient methodologies are needed to delineate the PTP functions. One strategy is to apply chemical genetic approaches utilizing potent and selective PTP inhibitors to study the physiological roles of the PTPs in vivo. Recent work using this approach to define the roles of PTP1B in insulin- and integrin-mediated processes is discussed. Another strategy is to apply activity-based proteomic techniques to measure globally PTP activity in both normal and pathological conditions. The ability to profile the entire PTP family on the basis of changes in their activity should greatly accelerate both the assignment of PTP function and the identification of potential therapeutic targets. Recent development on the design and characterization of activity-based PTP probes is highlighted.
Collapse
Affiliation(s)
- Zhong-Yin Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, USA.
| |
Collapse
|
49
|
De Dios I, Ramudo L, Alonso JR, Recio JS, Garcia-Montero AC, Manso MA. CD45 expression on rat acinar cells: Involvement in pro-inflammatory cytokine production. FEBS Lett 2005; 579:6355-60. [PMID: 16263122 DOI: 10.1016/j.febslet.2005.10.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Revised: 10/06/2005] [Accepted: 10/12/2005] [Indexed: 11/20/2022]
Abstract
CD45 transduces activation signals in inflammatory cells. We investigate CD45 expression on pancreatic acinar cells and examine its role in the inflammatory response which these cells have also shown under certain circumstances. Similar CD45 mRNA levels were found in acinar cells and leukocytes (positive control). Flow cytometric and immunohistochemical analysis showed a heterogeneous CD45 distribution on acinar cells. Activation of acinar cells by incubation with pancreatitis-associated ascitic fluid as evidencied by TNF-alpha production resulted in a decreased CD45 expression, suggesting that CD45 acts as a negative regulator of cytokine production. As a validation of this finding in vivo, a decrease in the acinar CD45 expression in parallel with an increased ability to produce TNF-alpha was found in rats with acute pancreatitis. Our data show that CD45 is constitutively expressed in acinar cells and suggest that it plays an important role in negatively regulating cytokine production.
Collapse
Affiliation(s)
- Isabel De Dios
- Department of Physiology and Pharmacology, University of Salamanca, Edificio Departamental, Campus Miguel de Unamuno, 37007 Salamanca, Spain.
| | | | | | | | | | | |
Collapse
|
50
|
Manger M, Scheck M, Prinz H, von Kries JP, Langer T, Saxena K, Schwalbe H, Fürstner A, Rademann J, Waldmann H. Discovery of Mycobacterium Tuberculosis Protein Tyrosine Phosphatase A (MptpA) Inhibitors Based on Natural Products and a Fragment-Based Approach. Chembiochem 2005; 6:1749-53. [PMID: 16196020 DOI: 10.1002/cbic.200500171] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Michael Manger
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|