1
|
Chen F, Zhao Y, Dai Y, Sun N, Gao X, Yin J, Zhou Z, Wu KJ. Chick Early Amniotic Fluid Alleviates Dextran-Sulfate-Sodium-Induced Colitis in Mice via T-Cell Receptor Pathway. Antioxidants (Basel) 2025; 14:51. [PMID: 39857385 PMCID: PMC11762673 DOI: 10.3390/antiox14010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/25/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic immune disease that is difficult to cure. We recently found that chick early amniotic fluid (ceAF) has notable anti-inflammatory and antioxidative properties, through its active components. This study demonstrates the potential of ceAF as a protective agent against UC. UPLC-MS mass spectrometry identified key components of ceAF, including various fatty acids and nucleosides. In vitro, ceAF improved viability in DSS-induced Caco-2 cells, reduced pro-inflammatory cytokines IL-1β and TNF-α, and increased the anti-inflammatory cytokine IL-10. It also upregulated the tight junction proteins ZO-1 and occludin. In DSS-induced UC mice, ceAF treatment alleviated weight loss, colon shortening, and disease activity, while improving histopathology, crypt depth, and colonic fibrosis. Mechanistically, ceAF's anti-inflammatory effects are mediated by inhibiting the overactivation of TCR signaling through the LCK/ZAP70/LAT pathway. Our findings suggest that ceAF could be a valuable nutritional intervention for UC, potentially enhancing existing functional foods aimed at managing this condition.
Collapse
Affiliation(s)
- Fan Chen
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi 214151, China; (F.C.); (X.G.); (J.Y.)
- Wuxi School of Medicine, Jiangnan University, Wuxi 214082, China; (Y.Z.); (Y.D.); (N.S.)
| | - Yining Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214082, China; (Y.Z.); (Y.D.); (N.S.)
| | - Yanfa Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi 214082, China; (Y.Z.); (Y.D.); (N.S.)
| | - Ning Sun
- Wuxi School of Medicine, Jiangnan University, Wuxi 214082, China; (Y.Z.); (Y.D.); (N.S.)
| | - Xuezheng Gao
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi 214151, China; (F.C.); (X.G.); (J.Y.)
| | - Jiajun Yin
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi 214151, China; (F.C.); (X.G.); (J.Y.)
| | - Zhenhe Zhou
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi 214151, China; (F.C.); (X.G.); (J.Y.)
| | - Ke-jia Wu
- Department of Psychiatry, The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi 214151, China; (F.C.); (X.G.); (J.Y.)
- Wuxi School of Medicine, Jiangnan University, Wuxi 214082, China; (Y.Z.); (Y.D.); (N.S.)
| |
Collapse
|
2
|
Noguchi R, Osaki J, Ono T, Adachi Y, Iwata S, Yoshimatsu Y, Sasaki K, Kawai A, Kondo T. Pharmacoproteogenomic approach identifies on-target kinase inhibitors for cancer drug repositioning. In Vitro Cell Dev Biol Anim 2024; 60:1200-1214. [PMID: 39422823 DOI: 10.1007/s11626-024-00983-3] [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: 07/02/2024] [Accepted: 09/01/2024] [Indexed: 10/19/2024]
Abstract
Drug repositioning of approved drugs offers advantages over de novo drug development for a rare type of cancer. To efficiently identify on-target drugs from clinically successful kinase inhibitors in cancer drug repositioning, drug screening and molecular profiling of cell lines are essential to exclude off-targets. We developed a pharmacoproteogenomic approach to identify on-target kinase inhibitors, combining molecular profiling of genomic features and kinase activity, and drug screening of patient-derived cell lines. This study examined eight patient-derived giant cell tumor of the bone (GCTB) cell lines, all of which harbored a signature mutation of H3-3A but otherwise without recurrent copy number variants and mutations. Kinase activity profiles of 100 tyrosine kinases with a three-dimensional substrate peptide array revealed that nine kinases were highly activated. Pharmacological screening of 60 clinically used kinase inhibitors found that nine drugs directed at 29 kinases strongly suppressed cell viability. We regarded ABL1, EGFR, and LCK as on-target kinases; among the two corresponding on-target kinase inhibitors, osimertinib and ponatinib emerged as on-target drugs whose target kinases were significantly activated. The remaining 26 kinases and seven kinase inhibitors were excluded as off-targets. Our pharmacoproteomic approach enabled the identification of on-target kinase inhibitors that are useful for drug repositioning.
Collapse
Affiliation(s)
- Rei Noguchi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Julia Osaki
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Takuya Ono
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yuki Adachi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Shuhei Iwata
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yuki Yoshimatsu
- Department of Patient-Derived Cancer Model, Tochigi Cancer Center, 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Kazuki Sasaki
- Department of Oncopeptidomics, Tochigi Cancer Center; 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology and Rehabilitation Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
3
|
Mok HL, Cheng KW, Xu Y, Huang C, Lyu C, Xu J, Hu D, Zhu L, Lin C, Tan HY, Bian Z. Modified Zhenwu Decoction suppresses chronic colitis via targeting macrophage CCR2/Fyn/p38 MAPK signaling axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155694. [PMID: 38733904 DOI: 10.1016/j.phymed.2024.155694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/15/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is associated with intestinal macrophage infiltration due to disruption of the mucosal barrier and bacterial invasion. Therefore, it is crucial to identify therapeutic agents capable of attenuating the macrophage-induced inflammatory response to preserve mucosal homeostasis and immune tolerance. The modified Zhenwu decoction (CDD-2103) is a novel herbal formulation developed based on the principles of Traditional Chinese medicine. To date, there are no clinically approved herbal formulations for UC with a well-known mechanism of action on macrophages. PURPOSE The objective of this study was to systematically investigate the inhibitory effect of the active fraction of CDD-2103 in a mouse model of chronic colitis and delineate the mechanisms underlying its inhibitory action. METHODS CDD-2103 was extracted into four fractions using organic solvents with increasing polarity. A chronic 49-day dextran sulfate sodium (DSS)-induced colitis mice model, closely resembling human clinical conditions, was used to examine the effect of CDD-2103 on chronic colitis. To confirm the effect of CDD-2103 on macrophages in this chronic colitis model, adoptive macrophage transfer and CCL2 supplementation were conducted. The mechanisms of action of CDD-2103 were further elucidated utilizing bone marrow-derived macrophages (BMDMs). Transcriptome analysis was conducted to gain insights into the underlying mechanism of action of CDD-2103 in BMDMs. RESULTS Our in vitro and in vivo findings demonstrated that the ethanol-enriched fraction of CDD-2103 exhibited significant anti-inflammatory effects, leading to the suppression of colitis severity. This effect was associated with diminished accumulation of colonic macrophages in the lamina propria of CDD-2103-intervened colitis mice. Specifically, CDD-2103 inhibited CCR2/L2-mediated proinflammatory macrophage infiltration into the colon without affecting macrophage proliferation. Mechanistically, CDD-2103 inhibited Fyn expression-mediated p38 MAPK activation and subsequently suppressed CCR2 expression in BMDMs. CONCLUSIONS Collectively, our study supports the potential use of CDD-2103 to limit macrophage infiltration, thereby reducing inflammation during UC treatment. CDD-2103 and the components in the ethanolic fraction are promising candidates for the development of novel drugs for UC management. Additionally, our study underscores Fyn-mediated CCR2 expression as a potential therapeutic target for the management of UC.
Collapse
Affiliation(s)
- Heung Lam Mok
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ka Wing Cheng
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China; School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yiqi Xu
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chunhua Huang
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China; School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Cheng Lyu
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jiaruo Xu
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China
| | - Die Hu
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China; School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Lin Zhu
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chengyuan Lin
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China
| | - Hor-Yue Tan
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China; School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| | - Zhaoxiang Bian
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China; School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
4
|
James J, Coelho A, Lahore GF, Hernandez CM, Forster F, Malissen B, Holmdahl R. Redox Regulation of LAT Enhances T Cell-Mediated Inflammation. Antioxidants (Basel) 2024; 13:499. [PMID: 38671946 PMCID: PMC11047684 DOI: 10.3390/antiox13040499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/04/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
The positional cloning of single nucleotide polymorphisms (SNPs) of the neutrophil cytosolic factor 1 (Ncf1) gene, advocating that a low oxidative burst drives autoimmune disease, demands an understanding of the underlying molecular causes. A cellular target could be T cells, which have been shown to be regulated by reactive oxygen species (ROS). However, the pathways by which ROS mediate T cell signaling remain unclear. The adaptor molecule linker for activation of T cells (LAT) is essential for coupling T cell receptor-mediated antigen recognition to downstream responses, and it contains several cysteine residues that have previously been suggested to be involved in redox regulation. To address the possibility that ROS regulate T cell-dependent inflammation through LAT, we established a mouse strain with cysteine-to-serine mutations at positions 120 and 172 (LATSS). We found that redox regulation of LAT through C120 and C172 mediate its localization and phosphorylation. LATSS mice had reduced numbers of double-positive thymocytes and naïve peripheral T cells. Importantly, redox insensitivity of LAT enhanced T cell-dependent autoimmune inflammation in collagen-induced arthritis (CIA), a mouse model of rheumatoid arthritis (RA). This effect was reversed on an NCF1-mutated (NCF1m1j), ROS-deficient, background. Overall, our data show that LAT is redox-regulated, acts to repress T cell activation, and is targeted by ROS induced by NCF1 in antigen-presenting cells (APCs).
Collapse
Affiliation(s)
- Jaime James
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; (J.J.); (A.C.); (G.F.L.); (C.M.H.); (F.F.)
| | - Ana Coelho
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; (J.J.); (A.C.); (G.F.L.); (C.M.H.); (F.F.)
| | - Gonzalo Fernandez Lahore
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; (J.J.); (A.C.); (G.F.L.); (C.M.H.); (F.F.)
| | - Clara M. Hernandez
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; (J.J.); (A.C.); (G.F.L.); (C.M.H.); (F.F.)
| | - Florian Forster
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; (J.J.); (A.C.); (G.F.L.); (C.M.H.); (F.F.)
| | - Bernard Malissen
- Centre d’Immunophénomique, Aix Marseille Université, INSERM, 13288 Marseille, France;
| | - Rikard Holmdahl
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; (J.J.); (A.C.); (G.F.L.); (C.M.H.); (F.F.)
| |
Collapse
|
5
|
Lee MS, Tuohy PJ, Kim CY, Yost PP, Lichauco K, Parrish HL, Van Doorslaer K, Kuhns MS. The CD4 transmembrane GGXXG and juxtamembrane (C/F)CV+C motifs mediate pMHCII-specific signaling independently of CD4-LCK interactions. eLife 2024; 12:RP88225. [PMID: 38639990 PMCID: PMC11031086 DOI: 10.7554/elife.88225] [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] [Indexed: 04/20/2024] Open
Abstract
CD4+ T cell activation is driven by five-module receptor complexes. The T cell receptor (TCR) is the receptor module that binds composite surfaces of peptide antigens embedded within MHCII molecules (pMHCII). It associates with three signaling modules (CD3γε, CD3δε, and CD3ζζ) to form TCR-CD3 complexes. CD4 is the coreceptor module. It reciprocally associates with TCR-CD3-pMHCII assemblies on the outside of a CD4+ T cells and with the Src kinase, LCK, on the inside. Previously, we reported that the CD4 transmembrane GGXXG and cytoplasmic juxtamembrane (C/F)CV+C motifs found in eutherian (placental mammal) CD4 have constituent residues that evolved under purifying selection (Lee et al., 2022). Expressing mutants of these motifs together in T cell hybridomas increased CD4-LCK association but reduced CD3ζ, ZAP70, and PLCγ1 phosphorylation levels, as well as IL-2 production, in response to agonist pMHCII. Because these mutants preferentially localized CD4-LCK pairs to non-raft membrane fractions, one explanation for our results was that they impaired proximal signaling by sequestering LCK away from TCR-CD3. An alternative hypothesis is that the mutations directly impacted signaling because the motifs normally play an LCK-independent role in signaling. The goal of this study was to discriminate between these possibilities. Using T cell hybridomas, our results indicate that: intracellular CD4-LCK interactions are not necessary for pMHCII-specific signal initiation; the GGXXG and (C/F)CV+C motifs are key determinants of CD4-mediated pMHCII-specific signal amplification; the GGXXG and (C/F)CV+C motifs exert their functions independently of direct CD4-LCK association. These data provide a mechanistic explanation for why residues within these motifs are under purifying selection in jawed vertebrates. The results are also important to consider for biomimetic engineering of synthetic receptors.
Collapse
Affiliation(s)
- Mark S Lee
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
| | - Peter J Tuohy
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
| | - Caleb Y Kim
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
| | - Philip P Yost
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
| | - Katrina Lichauco
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
| | - Heather L Parrish
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
| | - Koenraad Van Doorslaer
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
- School of Animal and Comparative Biomedical Sciences, The University of ArizonaTucsonUnited States
- Cancer Biology Graduate Interdisciplinary Program and Genetics Graduate Interdisciplinary Program, The University of ArizonaTucsonUnited States
- The BIO-5 Institute, The University of ArizonaTucsonUnited States
- The University of Arizona Cancer CenterTucsonUnited States
- The Arizona Center on Aging, The University of Arizona College of MedicineTucsonUnited States
| | - Michael S Kuhns
- Department of Immunobiology, The University of Arizona College of MedicineTucsonUnited States
- Cancer Biology Graduate Interdisciplinary Program and Genetics Graduate Interdisciplinary Program, The University of ArizonaTucsonUnited States
- The BIO-5 Institute, The University of ArizonaTucsonUnited States
- The University of Arizona Cancer CenterTucsonUnited States
- The Arizona Center on Aging, The University of Arizona College of MedicineTucsonUnited States
| |
Collapse
|
6
|
Lui VG, Hoenig M, Cabrera-Martinez B, Baxter RM, Garcia-Perez JE, Bailey O, Acharya A, Lundquist K, Capera J, Matusewicz P, Hartl FA, D’Abramo M, Alba J, Jacobsen EM, Niewolik D, Lorenz M, Pannicke U, Schulz AS, Debatin KM, Schamel WW, Minguet S, Gumbart JC, Dustin ML, Cambier JC, Schwarz K, Hsieh EW. A partial human LCK defect causes a T cell immunodeficiency with intestinal inflammation. J Exp Med 2024; 221:e20230927. [PMID: 37962568 PMCID: PMC10644909 DOI: 10.1084/jem.20230927] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 09/09/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Lymphocyte-specific protein tyrosine kinase (LCK) is essential for T cell antigen receptor (TCR)-mediated signal transduction. Here, we report two siblings homozygous for a novel LCK variant (c.1318C>T; P440S) characterized by T cell lymphopenia with skewed memory phenotype, infant-onset recurrent infections, failure to thrive, and protracted diarrhea. The patients' T cells show residual TCR signal transduction and proliferation following anti-CD3/CD28 and phytohemagglutinin (PHA) stimulation. We demonstrate in mouse models that complete (Lck-/-) versus partial (LckP440S/P440S) loss-of-function LCK causes disease with differing phenotypes. While both Lck-/- and LckP440S/P440S mice exhibit arrested thymic T cell development and profound T cell lymphopenia, only LckP440S/P440S mice show residual T cell proliferation, cytokine production, and intestinal inflammation. Furthermore, the intestinal disease in the LckP440S/P440S mice is prevented by CD4+ T cell depletion or regulatory T cell transfer. These findings demonstrate that P440S LCK spares sufficient T cell function to allow the maturation of some conventional T cells but not regulatory T cells-leading to intestinal inflammation.
Collapse
Affiliation(s)
- Victor G. Lui
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Manfred Hoenig
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Berenice Cabrera-Martinez
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ryan M. Baxter
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Josselyn E. Garcia-Perez
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Olivia Bailey
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Atanu Acharya
- School of Physics, Georgia Institute of Technology, Atlanta, GA, USA
- BioInspired Syracuse and Department of Chemistry, Syracuse University, Syracuse, NY, USA
| | - Karl Lundquist
- School of Physics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jesusa Capera
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Paul Matusewicz
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies and CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, University, Freiburg, Germany
| | - Frederike A. Hartl
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies and CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, University, Freiburg, Germany
| | - Marco D’Abramo
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | - Josephine Alba
- Department of Biology, Université de Fribourg, Fribourg, Switzerland
| | | | - Doris Niewolik
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Myriam Lorenz
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Ulrich Pannicke
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Ansgar S. Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | | | - Wolfgang W. Schamel
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies and CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, University, Freiburg, Germany
| | - Susana Minguet
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies and CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, University, Freiburg, Germany
| | - James C. Gumbart
- School of Physics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael L. Dustin
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - John C. Cambier
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Human Immunology and Immunotherapy Initiative, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| | - Klaus Schwarz
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service Baden-Wuerttemberg-Hessen, Ulm, Germany
| | - Elena W.Y. Hsieh
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Human Immunology and Immunotherapy Initiative, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
- Department of Pediatrics, Section of Allergy and Immunology, Children’s Hospital Colorado, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| |
Collapse
|
7
|
Lanz AL, Erdem S, Ozcan A, Ceylaner G, Cansever M, Ceylaner S, Conca R, Magg T, Acuto O, Latour S, Klein C, Patiroglu T, Unal E, Eken A, Hauck F. A Novel Biallelic LCK Variant Resulting in Profound T-Cell Immune Deficiency and Review of the Literature. J Clin Immunol 2023; 44:1. [PMID: 38100037 PMCID: PMC10724324 DOI: 10.1007/s10875-023-01602-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 10/06/2023] [Indexed: 12/18/2023]
Abstract
Lymphocyte-specific protein tyrosine kinase (LCK) is an SRC-family kinase critical for initiation and propagation of T-cell antigen receptor (TCR) signaling through phosphorylation of TCR-associated CD3 chains and recruited downstream molecules. Until now, only one case of profound T-cell immune deficiency with complete LCK deficiency [1] caused by a biallelic missense mutation (c.1022T>C, p.L341P) and three cases of incomplete LCK deficiency [2] caused by a biallelic splice site mutation (c.188-2A>G) have been described. Additionally, deregulated LCK expression has been associated with genetically undefined immune deficiencies and hematological malignancies. Here, we describe the second case of complete LCK deficiency in a 6-month-old girl born to consanguineous parents presenting with profound T-cell immune deficiency. Whole exome sequencing (WES) revealed a novel pathogenic biallelic missense mutation in LCK (c.1393T>C, p.C465R), which led to the absence of LCK protein expression and phosphorylation, and a consecutive decrease in proximal TCR signaling. Loss of conventional CD4+ and CD8+ αβT-cells and homeostatic T-cell expansion was accompanied by increased γδT-cell and Treg percentages. Surface CD4 and CD8 co-receptor expression was reduced in the patient T-cells, while the heterozygous mother had impaired CD4 and CD8 surface expression to a lesser extent. We conclude that complete LCK deficiency is characterized by profound T-cell immune deficiency, reduced CD4 and CD8 surface expression, and a characteristic TCR signaling disorder. CD4 and CD8 surface expression may be of value for early detection of mono- and/or biallelic LCK deficiency.
Collapse
Affiliation(s)
- Anna-Lisa Lanz
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Lindwurmstrasse 4, 80337, Munich, Germany
| | - Serife Erdem
- Department of Medical Biology, Faculty of Medicine, Erciyes University, 38030, Kayseri, Turkey
- Molecular Biology and Genetics Department, Gevher Nesibe Genome and Stem Cell Institute, Betul-Ziya Eren Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| | - Alper Ozcan
- Molecular Biology and Genetics Department, Gevher Nesibe Genome and Stem Cell Institute, Betul-Ziya Eren Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| | | | - Murat Cansever
- Division of Pediatric Hematology & Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | | | - Raffaele Conca
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Lindwurmstrasse 4, 80337, Munich, Germany
| | - Thomas Magg
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Lindwurmstrasse 4, 80337, Munich, Germany
| | - Oreste Acuto
- T Cell Signalling Laboratory, Sir William Dunn School of Pathology, Oxford University, Oxford, OX2 3RE, UK
| | - Sylvain Latour
- Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, INSERM UMR1163, Paris, France
| | - Christoph Klein
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Lindwurmstrasse 4, 80337, Munich, Germany
| | - Turkan Patiroglu
- Division of Pediatric Hematology & Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Ekrem Unal
- Molecular Biology and Genetics Department, Gevher Nesibe Genome and Stem Cell Institute, Betul-Ziya Eren Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
- Intergen, Ankara, Turkey
- Hasan Kalyoncu University, Faculty of Health Sciences, Medical Point Hospital, Gaziantep, Türkiye
| | - Ahmet Eken
- Department of Medical Biology, Faculty of Medicine, Erciyes University, 38030, Kayseri, Turkey.
- Molecular Biology and Genetics Department, Gevher Nesibe Genome and Stem Cell Institute, Betul-Ziya Eren Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey.
| | - Fabian Hauck
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Lindwurmstrasse 4, 80337, Munich, Germany.
- Munich Centre for Rare Diseases (M-ZSELMU), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
8
|
Lee MS, Tuohy PJ, Kim CY, Yost P, Lichauco K, Parrish HL, Van Doorslaer K, Kuhns MS. The CD4 transmembrane GGXXG and juxtamembrane (C/F)CV+C motifs mediate pMHCII-specific signaling independently of CD4-LCK interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539613. [PMID: 37214965 PMCID: PMC10197521 DOI: 10.1101/2023.05.05.539613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
CD4+ T cell activation is driven by 5-module receptor complexes. The T cell receptor (TCR) is the receptor module that binds composite surfaces of peptide antigens embedded within MHCII molecules (pMHCII). It associates with three signaling modules (CD3γε, CD3δε, and CD3ζζ) to form TCR-CD3 complexes. CD4 is the coreceptor module. It reciprocally associates with TCR-CD3-pMHCII assemblies on the outside of a CD4+ T cells and with the Src kinase, LCK, on the inside. Previously, we reported that the CD4 transmembrane GGXXG and cytoplasmic juxtamembrane (C/F)CV+C motifs found in eutherian (placental mammal) CD4 have constituent residues that evolved under purifying selection (Lee, et al., 2022). Expressing mutants of these motifs together in T cell hybridomas increased CD4-LCK association but reduced CD3ζ, ZAP70, and PLCγ1 phosphorylation levels, as well as IL-2 production, in response to agonist pMHCII. Because these mutants preferentially localized CD4-LCK pairs to non-raft membrane fractions, one explanation for our results was that they impaired proximal signaling by sequestering LCK away from TCR-CD3. An alternative hypothesis is that the mutations directly impacted signaling because the motifs normally play an LCK-independent role in signaling. The goal of this study was to discriminate between these possibilities. Using T cell hybridomas, our results indicate that: intracellular CD4-LCK interactions are not necessary for pMHCII-specific signal initiation; the GGXXG and (C/F)CV+C motifs are key determinants of CD4-mediated pMHCII-specific signal amplification; the GGXXG and (C/F)CV+C motifs exert their functions independently of direct CD4-LCK association. These data provide a mechanistic explanation for why residues within these motifs are under purifying selection in jawed vertebrates. The results are also important to consider for biomimetic engineering of synthetic receptors.
Collapse
Affiliation(s)
- Mark S Lee
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Peter J Tuohy
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Caleb Y Kim
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Philip Yost
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Katrina Lichauco
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Heather L Parrish
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Koenraad Van Doorslaer
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ 85724, USA
- Cancer Biology Graduate Interdisciplinary Program and Genetics Graduate Interdisciplinary Program, The University of Arizona, Tucson, AZ 85724, USA
- The BIO-5 Institute, The University of Arizona, Tucson, AZ 85724, USA
- The University of Arizona Cancer Center, Tucson, AZ, USA
- The Arizona Center on Aging, The University of Arizona College of Medicine, Tucson, AZ, USA
| | - Michael S Kuhns
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724, USA
- Cancer Biology Graduate Interdisciplinary Program and Genetics Graduate Interdisciplinary Program, The University of Arizona, Tucson, AZ 85724, USA
- The BIO-5 Institute, The University of Arizona, Tucson, AZ 85724, USA
- The University of Arizona Cancer Center, Tucson, AZ, USA
- The Arizona Center on Aging, The University of Arizona College of Medicine, Tucson, AZ, USA
| |
Collapse
|
9
|
Tsubata T. Siglec cis-ligands and their roles in the immune system. Glycobiology 2023; 33:532-544. [PMID: 37154567 DOI: 10.1093/glycob/cwad038] [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: 10/20/2022] [Revised: 04/14/2023] [Indexed: 05/10/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectins are a family of membrane molecules primarily expressed in immune cells. Most of them are inhibitory receptors containing immunoreceptor tyrosine-based inhibition motifs in the cytoplasmic tail. On the cell surface, sialic acid-binding immunoglobulin-like lectins are mostly bound by sialylated glycans on membrane molecules expressed in the same cell (cis-ligands). Although ligands of sialic acid-binding immunoglobulin-like lectins are not efficiently identified by conventional methods such as immunoprecipitation, in situ labeling including proximity labeling is useful in identifying both cis-ligands and the sialylated ligands expressed by other cells (trans-ligands) of sialic acid-binding immunoglobulin-like lectins. Interaction of the inhibitory sialic acid-binding immunoglobulin-like lectins with cis-ligands including both those with and without signaling function modulates the inhibitory activity of sialic acid-binding immunoglobulin-like lectins by multiple different ways. This interaction also modulates signaling function of the cis-ligands. So far, little is known about the role of the interaction between sialic acid-binding immunoglobulin-like lectins and the cis-ligands. Nonetheless, recent studies showed that the inhibitory activity of CD22 (also known as Siglec-2) is regulated by endogenous ligands, most likely cis-ligands, differentially in resting B cells and those in which B-cell antigen receptor is ligated. This differential regulation plays a role in quality control of signaling-competent B cells and also partial restoration of B-cell antigen receptor signaling in immunodeficient B cells.
Collapse
Affiliation(s)
- Takeshi Tsubata
- Department of Pathology, Nihon University School of Dentistry, Tokyo 101-8310, Japan
| |
Collapse
|
10
|
Wang A, Wang J, Mao M, Zhao X, Li Q, Xuan R, Li F, Chao T. Analyses of lncRNAs, circRNAs, and the Interactions between ncRNAs and mRNAs in Goat Submandibular Glands Reveal Their Potential Function in Immune Regulation. Genes (Basel) 2023; 14:187. [PMID: 36672927 PMCID: PMC9859278 DOI: 10.3390/genes14010187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/01/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
As part of one of the main ruminants, goat salivary glands hardly secrete digestive enzymes, but play an important role in immunity. The immune function of goat salivary glands significantly changes with age, while the expression profile and specific function of non-coding RNA during this process are unknown. In this study, transcriptome sequencing was performed on submandibular gland (SMG) tissues of 1-month-old, 12-month-old, and 24-month-old goats, revealing the expression patterns of lncRNA and circRNA at different ages. A total of 369 lncRNAs and 1699 circRNAs were found to be differentially expressed. Functional enrichment analyses showed that the lncRNA regulated target mRNAs and circRNA host genes were significantly enriched in immune-related GO terms and pathways. CeRNA network analysis showed that the key differentially expressed circRNAs and lncRNAs mainly regulate the key immune-related genes ITGB2, LCP2, PTPRC, SYK, and ZAP70 through competitive binding with miR-141-x, miR-29-y, and chi-miR-29b-3p, thereby affecting the natural killer cell-mediated cytotoxicity pathway, the T cell receptor signaling pathway, and other immune-related pathways. It should be noted that the expression of key circRNAs, lncRNAs, and key immune-related genes in goat SMGs decreased significantly with the growth of the goat. This is the first reporting of lncRNAs, circRNAs, and ceRNA network regulation in goat SMGs. Our study contributes to the knowledge of changes in the expression of non-coding RNAs during SMG development in goats and provides new insights into the relationship between non-coding RNAs and salivary gland immune function in goats.
Collapse
Affiliation(s)
- Aili Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271000, China
| | - Jianmin Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271000, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian 271000, China
| | - Meina Mao
- Shandong Peninsula Engineering Research Center of Comprehensive Brine Utilization, Weifang University of Science and Technology, Shouguang 262700, China
| | - Xiaodong Zhao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271000, China
- Shandong Vocational Animal Science and Veterinary College, Weifang 261000, China
| | - Qing Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271000, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian 271000, China
| | - Rong Xuan
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271000, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian 271000, China
| | - Fajun Li
- Shandong Peninsula Engineering Research Center of Comprehensive Brine Utilization, Weifang University of Science and Technology, Shouguang 262700, China
| | - Tianle Chao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271000, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian 271000, China
| |
Collapse
|
11
|
Unique roles of co-receptor-bound LCK in helper and cytotoxic T cells. Nat Immunol 2023; 24:174-185. [PMID: 36564464 PMCID: PMC9810533 DOI: 10.1038/s41590-022-01366-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 10/20/2022] [Indexed: 12/24/2022]
Abstract
The kinase LCK and CD4/CD8 co-receptors are crucial components of the T cell antigen receptor (TCR) signaling machinery, leading to key T cell fate decisions. Despite decades of research, the roles of CD4-LCK and CD8-LCK interactions in TCR triggering in vivo remain unknown. In this study, we created animal models expressing endogenous levels of modified LCK to resolve whether and how co-receptor-bound LCK drives TCR signaling. We demonstrated that the role of LCK depends on the co-receptor to which it is bound. The CD8-bound LCK is largely dispensable for antiviral and antitumor activity of cytotoxic T cells in mice; however, it facilitates CD8+ T cell responses to suboptimal antigens in a kinase-dependent manner. By contrast, the CD4-bound LCK is required for efficient development and function of helper T cells via a kinase-independent stabilization of surface CD4. Overall, our findings reveal the role of co-receptor-bound LCK in T cell biology, show that CD4- and CD8-bound LCK drive T cell development and effector immune responses using qualitatively different mechanisms and identify the co-receptor-LCK interactions as promising targets for immunomodulation.
Collapse
|
12
|
Laukkanen S, Veloso A, Yan C, Oksa L, Alpert EJ, Do D, Hyvärinen N, McCarthy K, Adhikari A, Yang Q, Iyer S, Garcia SP, Pello A, Ruokoranta T, Moisio S, Adhikari S, Yoder JA, Gallagher K, Whelton L, Allen JR, Jin AH, Loontiens S, Heinäniemi M, Kelliher M, Heckman CA, Lohi O, Langenau DM. Therapeutic targeting of LCK tyrosine kinase and mTOR signaling in T-cell acute lymphoblastic leukemia. Blood 2022; 140:1891-1906. [PMID: 35544598 PMCID: PMC10082361 DOI: 10.1182/blood.2021015106] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/19/2022] [Indexed: 11/20/2022] Open
Abstract
Relapse and refractory T-cell acute lymphoblastic leukemia (T-ALL) has a poor prognosis, and new combination therapies are sorely needed. Here, we used an ex vivo high-throughput screening platform to identify drug combinations that kill zebrafish T-ALL and then validated top drug combinations for preclinical efficacy in human disease. This work uncovered potent drug synergies between AKT/mTORC1 (mammalian target of rapamycin complex 1) inhibitors and the general tyrosine kinase inhibitor dasatinib. Importantly, these same drug combinations effectively killed a subset of relapse and dexamethasone-resistant zebrafish T-ALL. Clinical trials are currently underway using the combination of mTORC1 inhibitor temsirolimus and dasatinib in other pediatric cancer indications, leading us to prioritize this therapy for preclinical testing. This combination effectively curbed T-ALL growth in human cell lines and primary human T-ALL and was well tolerated and effective in suppressing leukemia growth in patient-derived xenografts (PDX) grown in mice. Mechanistically, dasatinib inhibited phosphorylation and activation of the lymphocyte-specific protein tyrosine kinase (LCK) to blunt the T-cell receptor (TCR) signaling pathway, and when complexed with mTORC1 inhibition, induced potent T-ALL cell killing through reducing MCL-1 protein expression. In total, our work uncovered unexpected roles for the LCK kinase and its regulation of downstream TCR signaling in suppressing apoptosis and driving continued leukemia growth. Analysis of a wide array of primary human T-ALLs and PDXs grown in mice suggest that combination of temsirolimus and dasatinib treatment will be efficacious for a large fraction of human T-ALLs.
Collapse
Affiliation(s)
- Saara Laukkanen
- Tampere Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Alexandra Veloso
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Chuan Yan
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Laura Oksa
- Tampere Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Eric J. Alpert
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Daniel Do
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Noora Hyvärinen
- Tampere Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Karin McCarthy
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Abhinav Adhikari
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Qiqi Yang
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Sowmya Iyer
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Sara P. Garcia
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Annukka Pello
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Tanja Ruokoranta
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Sanni Moisio
- The Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Sadiksha Adhikari
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Jeffrey A. Yoder
- Department of Molecular Biomedical Sciences, Comparative Medicine Institute, and Center for Human Health and the Environment, North Carolina State University, Raleigh, NC
| | - Kayleigh Gallagher
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Lauren Whelton
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - James R. Allen
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Alex H. Jin
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Siebe Loontiens
- Cancer Research Institute Ghent and Center for Medical Genetics, Ghent, Belgium
| | - Merja Heinäniemi
- The Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Michelle Kelliher
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Caroline A. Heckman
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Olli Lohi
- Tampere Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere University Hospital, Tays Cancer Center, Tampere, Finland
| | - David M. Langenau
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
13
|
Ding C, Xu H, Yu Z, Roulis M, Qu R, Zhou J, Oh J, Crawford J, Gao Y, Jackson R, Sefik E, Li S, Wei Z, Skadow M, Yin Z, Ouyang X, Wang L, Zou Q, Su B, Hu W, Flavell RA, Li HB. RNA m 6A demethylase ALKBH5 regulates the development of γδ T cells. Proc Natl Acad Sci U S A 2022; 119:e2203318119. [PMID: 35939687 PMCID: PMC9388086 DOI: 10.1073/pnas.2203318119] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
γδ T cells are an abundant T cell population at the mucosa and are important in providing immune surveillance as well as maintaining tissue homeostasis. However, despite γδ T cells' origin in the thymus, detailed mechanisms regulating γδ T cell development remain poorly understood. N6-methyladenosine (m6A) represents one of the most common posttranscriptional modifications of messenger RNA (mRNA) in mammalian cells, but whether it plays a role in γδ T cell biology is still unclear. Here, we show that depletion of the m6A demethylase ALKBH5 in lymphocytes specifically induces an expansion of γδ T cells, which confers enhanced protection against gastrointestinal Salmonella typhimurium infection. Mechanistically, loss of ALKBH5 favors the development of γδ T cell precursors by increasing the abundance of m6A RNA modification in thymocytes, which further reduces the expression of several target genes including Notch signaling components Jagged1 and Notch2. As a result, impairment of Jagged1/Notch2 signaling contributes to enhanced proliferation and differentiation of γδ T cell precursors, leading to an expanded mature γδ T cell repertoire. Taken together, our results indicate a checkpoint role of ALKBH5 and m6A modification in the regulation of γδ T cell early development.
Collapse
Affiliation(s)
- Chenbo Ding
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
| | - Hao Xu
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Zhibin Yu
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
| | - Manolis Roulis
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Rihao Qu
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
- dProgram of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520
- eDepartment of Pathology, Yale University School of Medicine, New Haven, CT 06510
| | - Jing Zhou
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
| | - Joonseok Oh
- fDepartment of Chemistry, Yale University, New Haven, CT 06520
- gChemical Biology Institute, Yale University, West Haven, CT 06516
| | - Jason Crawford
- fDepartment of Chemistry, Yale University, New Haven, CT 06520
- gChemical Biology Institute, Yale University, West Haven, CT 06516
- hDepartment of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06520
| | - Yimeng Gao
- iSection of Hematology, Yale Cancer Center and Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
- jYale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06520
- kYale RNA Center, Yale University School of Medicine, New Haven, CT 06520
| | - Ruaidhrí Jackson
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Esen Sefik
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Simiao Li
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Zheng Wei
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Mathias Skadow
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Zhinan Yin
- lZhuhai Precision Medical Center, Zhuhai People’s Hospital, Jinan University, Zhuhai 519000, Guangdong, China
- mBiomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China
| | - Xinshou Ouyang
- nSection of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Lei Wang
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qiang Zou
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bing Su
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
| | - Weiguo Hu
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- 2To whom correspondence may be addressed. , , or
| | - Richard A. Flavell
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
- oHHMI, Yale University School of Medicine, New Haven, CT 06520
- 2To whom correspondence may be addressed. , , or
| | - Hua-Bing Li
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
- 2To whom correspondence may be addressed. , , or
| |
Collapse
|
14
|
Kästle M, Merten C, Hartig R, Plaza-Sirvent C, Schmitz I, Bommhardt U, Schraven B, Simeoni L. Type of PaperY192 within the SH2 Domain of Lck Regulates TCR Signaling Downstream of PLC-γ1 and Thymic Selection. Int J Mol Sci 2022; 23:ijms23137271. [PMID: 35806279 PMCID: PMC9267008 DOI: 10.3390/ijms23137271] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Signaling via the TCR, which is initiated by the Src-family tyrosine kinase Lck, is crucial for the determination of cell fates in the thymus. Because of its pivotal role, ablation of Lck results in a profound block of T-cell development. Here, we show that, in addition to its well-known function in the initiation of TCR signaling, Lck also acts at a more downstream level. This novel function of Lck is determined by the tyrosine residue (Y192) located in its SH2 domain. Thymocytes from knock-in mice expressing a phosphomimetic Y192E mutant of Lck initiate TCR signaling upon CD3 cross-linking up to the level of PLC-γ1 phosphorylation. However, the activation of downstream pathways including Ca2+ influx and phosphorylation of Erk1/2 are impaired. Accordingly, positive and negative selections are blocked in LckY192E knock-in mice. Collectively, our data indicate that Lck has a novel function downstream of PLCγ-1 in the regulation of thymocyte differentiation and selection.
Collapse
Affiliation(s)
- Matthias Kästle
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
| | - Camilla Merten
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
| | - Roland Hartig
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
| | - Carlos Plaza-Sirvent
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
- Department of Molecular Immunology, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Ingo Schmitz
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
- Department of Molecular Immunology, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Ursula Bommhardt
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
- Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
- Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke University, 39120 Magdeburg, Germany
- Correspondence: (B.S.); (L.S.)
| | - Luca Simeoni
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.K.); (C.M.); (R.H.); (C.P.-S.); (I.S.); (U.B.)
- Health Campus Immunology, Infectiology and Inflammation (GC-I3), Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke University, 39120 Magdeburg, Germany
- Correspondence: (B.S.); (L.S.)
| |
Collapse
|
15
|
Zhang Z, He Y, Lin R, Lan J, Fan Y, Wang P, Jia C. Identification of Important Modules and Biomarkers That Are Related to Immune Infiltration Cells in Severe Burns Based on Weighted Gene Co-Expression Network Analysis. Front Genet 2022; 13:908510. [PMID: 35754830 PMCID: PMC9218676 DOI: 10.3389/fgene.2022.908510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Immunosuppression is an important trigger for infection and a significant cause of death in patients with severe burns. Nevertheless, the prognostic value of immune-related genes remains unclear. This study aimed to identify the biomarkers related to immunosuppression in severe burns. Methods: The gene expression profile and clinical data of 185 burn and 75 healthy samples were obtained from the GEO database. Immune infiltration analysis and gene set variation analysis were utilized to identify the disorder of circulating immune cells. A weighted gene co-expression network analysis (WGCNA) was carried out to select immune-related gene modules. Enrichment analysis and protein-protein interaction (PPI) network were performed to select hub genes. Next, LASSO and logistic regression were utilized to construct the hazard regression model with a survival state. Finally, we investigated the correlation between high- and low-risk patients in total burn surface area (TBSA), age, and inhalation injury. Results: Gene set variation analysis (GSVA) and immune infiltration analysis showed that neutrophils increased and T cells decreased in severe burns. In WGCNA, four modular differently expressed in burns and controls were related to immune cells. Based on PPI and enrichment analysis, 210 immune-related genes were identified, mainly involved in T-cell inhibition and neutrophil activation. In LASSO and logistic regression, we screened out key genes, including LCK, SKAP1 and GZMB, and LY9. In the ROC analysis, the area under the curve (AUC) of key genes was 0.945, indicating that the key genes had excellent diagnostic value. Finally, we discovered that the key genes were related to T cells, and the regression model performed well when accompanied by TBSA and age. Conclusion: We identified LCK, SKAP1, GZMB, and LY9 as good prognostic biomarkers that may play a role in post-burn immunosuppression against T-cell dysfunction and as potential immunotherapeutic targets for transformed T-cell dysfunction.
Collapse
Affiliation(s)
- Zexin Zhang
- Department of Burns and Plastic and Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yan He
- Department of Burns and Plastic and Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Rongjie Lin
- Department of Orthopedics, The 900th Hospital of Joint Logistic Support Force, Fuzhou, China
| | - Junhong Lan
- Department of Burns and Plastic and Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yueying Fan
- Department of Burns and Plastic and Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Peng Wang
- Department of Burns and Plastic and Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.,Department of Burns and Plastic and Cosmetic Surgery, The Ninth Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chiyu Jia
- Department of Burns and Plastic and Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
16
|
Huseby ES, Teixeiro E. The perception and response of T cells to a changing environment are based on the law of initial value. Sci Signal 2022; 15:eabj9842. [PMID: 35639856 PMCID: PMC9290192 DOI: 10.1126/scisignal.abj9842] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
αβ T cells are critical components of the adaptive immune system and are capable of inducing sterilizing immunity after pathogen infection and eliminating transformed tumor cells. The development and function of T cells are controlled through the T cell antigen receptor, which recognizes peptides displayed on major histocompatibility complex (MHC) molecules. Here, we review how T cells generate the ability to recognize self-peptide-bound MHC molecules and use signals derived from these interactions to instruct cellular development, activation thresholds, and functional specialization in the steady state and during immune responses. We argue that the basic tenants of T cell development and function follow Weber-Fetcher's law of just noticeable differences and Wilder's law of initial value. Together, these laws argue that the ability of a system to respond and the quality of that response are scalable to the basal state of that system. Manifestation of these laws in T cells generates clone-specific activation thresholds that are based on perceivable differences between homeostasis and pathogen encounter (self versus nonself discrimination), as well as poised states for subsequent differentiation into specific effector cell lineages.
Collapse
Affiliation(s)
- Eric S. Huseby
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
17
|
Acute Csk inhibition hinders B cell activation by constraining the PI3 kinase pathway. Proc Natl Acad Sci U S A 2021; 118:2108957118. [PMID: 34675079 PMCID: PMC8639343 DOI: 10.1073/pnas.2108957118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2021] [Indexed: 12/30/2022] Open
Abstract
B lymphocytes recognize pathogenic antigens and become activated via their B cell receptors (BCR). This BCR-dependent activation is controlled by Src-family kinases (SFKs). It is unclear how B cells tolerate the fluctuations of SFK activities and maintain unresponsiveness in the absence of foreign antigens. Using a chemical-genetic system, we acutely inhibited C-terminal Src kinase to enhance the SFK activity in mouse B cells. Surprisingly, we observed marked inhibition of BCR-downstream signaling due to associated impairment of the phosphatidylinositol-trisphosphate pathway. These results reveal the critical importance of maintaining a proper amount of SFK activity in quiescent B cells for appropriate BCR-dependent responses, which may be critical for naïve B cell unresponsiveness to self-antigens to maintain peripheral tolerance. T cell antigen receptor (TCR) and B cell antigen receptor (BCR) signaling are initiated and tightly regulated by Src-family kinases (SFKs). SFKs positively regulate TCR signaling in naïve T cells but have both positive and negative regulatory roles in BCR signaling in naïve B cells. The proper regulation of their activities depends on the opposing actions of receptor tyrosine phosphatases CD45 and CD148 and the cytoplasmic tyrosine kinase C-terminal Src kinase Csk. Csk is a major negative regulator of SFKs. Using a PP1-analog-sensitive Csk (CskAS) system, we have previously shown that inhibition of CskAS increases SFK activity, leading to augmentation of responses to weak TCR stimuli in T cells. However, the effects of Csk inhibition in B cells were not known. In this study, we surprisingly found that inhibition of CskAS led to marked inhibition of BCR-stimulated cytoplasmic free calcium increase and Erk activation despite increased SFK activation in B cells, contrasting the effects observed in T cells. Further investigation revealed that acute CskAS inhibition suppressed BCR-mediated phosphatidylinositol 3,4,5-trisphosphate (PIP3) production in B cells. Restoring PIP3 levels in B cells by CD19 cross-linking or SHIP1 deficiency eliminated the negative regulatory effect of CskAS inhibition. This reveals the critical role of Csk in maintaining an appropriate level of SFK activity and regulating PIP3 amounts as a means of compensating for SFK fluctuations to prevent inappropriate B cell activation. This regulatory mechanism controlling PIP3 amounts may also contribute to B cell anergy and self-tolerance.
Collapse
|
18
|
Guglietti B, Sivasankar S, Mustafa S, Corrigan F, Collins-Praino LE. Fyn Kinase Activity and Its Role in Neurodegenerative Disease Pathology: a Potential Universal Target? Mol Neurobiol 2021; 58:5986-6005. [PMID: 34432266 DOI: 10.1007/s12035-021-02518-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022]
Abstract
Fyn is a non-receptor tyrosine kinase belonging to the Src family of kinases (SFKs) which has been implicated in several integral functions throughout the central nervous system (CNS), including myelination and synaptic transmission. More recently, Fyn dysfunction has been associated with pathological processes observed in neurodegenerative diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD) and Parkinson's disease (PD). Neurodegenerative diseases are amongst the leading cause of death and disability worldwide and, due to the ageing population, prevalence is predicted to rise in the coming years. Symptoms across neurodegenerative diseases are both debilitating and degenerative in nature and, concerningly, there are currently no disease-modifying therapies to prevent their progression. As such, it is important to identify potential new therapeutic targets. This review will outline the role of Fyn in normal/homeostatic processes, as well as degenerative/pathological mechanisms associated with neurodegenerative diseases, such as demyelination, pathological protein aggregation, neuroinflammation and cognitive dysfunction.
Collapse
Affiliation(s)
- Bianca Guglietti
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Srisankavi Sivasankar
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Sanam Mustafa
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia
| | - Frances Corrigan
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Lyndsey E Collins-Praino
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia. .,ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
19
|
Kent A, Longino NV, Christians A, Davila E. Naturally Occurring Genetic Alterations in Proximal TCR Signaling and Implications for Cancer Immunotherapy. Front Immunol 2021; 12:658611. [PMID: 34012443 PMCID: PMC8126620 DOI: 10.3389/fimmu.2021.658611] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
T cell-based immunotherapies including genetically engineered T cells, adoptive transfer of tumor-infiltrating lymphocytes, and immune checkpoint blockade highlight the impressive anti-tumor effects of T cells. These successes have provided new hope to many cancer patients with otherwise poor prognoses. However, only a fraction of patients demonstrates durable responses to these forms of therapies and many develop significant immune-mediated toxicity. These heterogeneous clinical responses suggest that underlying nuances in T cell genetics, phenotypes, and activation states likely modulate the therapeutic impact of these approaches. To better characterize known genetic variations that may impact T cell function, we 1) review the function of early T cell receptor-specific signaling mediators, 2) offer a synopsis of known mutations and genetic alterations within the associated molecules, 3) discuss the link between these mutations and human disease and 4) review therapeutic strategies under development or in clinical testing that target each of these molecules for enhancing anti-tumor T cell activity. Finally, we discuss novel engineering approaches that could be designed based on our understanding of the function of these molecules in health and disease.
Collapse
Affiliation(s)
- Andrew Kent
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative, University of Colorado, Aurora, CO, United States
- University of Colorado Comprehensive Cancer Center, Aurora, CO, United States
| | - Natalie V. Longino
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative, University of Colorado, Aurora, CO, United States
- University of Colorado Comprehensive Cancer Center, Aurora, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Allison Christians
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative, University of Colorado, Aurora, CO, United States
- University of Colorado Comprehensive Cancer Center, Aurora, CO, United States
| | - Eduardo Davila
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative, University of Colorado, Aurora, CO, United States
- University of Colorado Comprehensive Cancer Center, Aurora, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| |
Collapse
|
20
|
van der Donk LEH, Ates LS, van der Spek J, Tukker LM, Geijtenbeek TBH, van Heijst JWJ. Separate signaling events control TCR downregulation and T cell activation in primary human T cells. IMMUNITY INFLAMMATION AND DISEASE 2020; 9:223-238. [PMID: 33350598 PMCID: PMC7860602 DOI: 10.1002/iid3.383] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/27/2020] [Accepted: 11/17/2020] [Indexed: 12/02/2022]
Abstract
Introduction T‐cell antigen receptor (TCR) interaction with cognate peptide:MHC complexes trigger clustering of TCR:CD3 complexes and signal transduction. Triggered TCR:CD3 complexes are rapidly internalized and degraded in a process called ligand‐induced TCR downregulation. Classic studies in immortalized T‐cell lines have revealed a major role for the Src family kinase Lck in TCR downregulation. However, to what extent a similar mechanism operates in primary human T cells remains unclear. Methods Here, we developed an anti‐CD3‐mediated TCR downregulation assay, in which T‐cell gene expression in primary human T cells can be knocked down by microRNA constructs. In parallel, we used CRISPR/Cas9‐mediated knockout in Jurkat cells for validation experiments. Results We efficiently knocked down the expression of tyrosine kinases Lck, Fyn, and ZAP70, and found that, whereas this impaired T cell activation and effector function, TCR downregulation was not affected. Although TCR downregulation was marginally inhibited by the simultaneous knockdown of Lck and Fyn, its full abrogation required broad‐acting tyrosine kinase inhibitors. Conclusions These data suggest that there is substantial redundancy in the contribution of individual tyrosine kinases to TCR downregulation in primary human T cells. Our results highlight that TCR downregulation and T cell activation are controlled by different signaling events and illustrate the need for further research to untangle these processes.
Collapse
Affiliation(s)
- Lieve E H van der Donk
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Louis S Ates
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jet van der Spek
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Laura M Tukker
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeroen W J van Heijst
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Neogene Therapeutics, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Mørch AM, Bálint Š, Santos AM, Davis SJ, Dustin ML. Coreceptors and TCR Signaling - the Strong and the Weak of It. Front Cell Dev Biol 2020; 8:597627. [PMID: 33178706 PMCID: PMC7596257 DOI: 10.3389/fcell.2020.597627] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 09/28/2020] [Indexed: 12/02/2022] Open
Abstract
The T-cell coreceptors CD4 and CD8 have well-characterized and essential roles in thymic development, but how they contribute to immune responses in the periphery is unclear. Coreceptors strengthen T-cell responses by many orders of magnitude - beyond a million-fold according to some estimates - but the mechanisms underlying these effects are still debated. T-cell receptor (TCR) triggering is initiated by the binding of the TCR to peptide-loaded major histocompatibility complex (pMHC) molecules on the surfaces of other cells. CD4 and CD8 are the only T-cell proteins that bind to the same pMHC ligand as the TCR, and can directly associate with the TCR-phosphorylating kinase Lck. At least three mechanisms have been proposed to explain how coreceptors so profoundly amplify TCR signaling: (1) the Lck recruitment model and (2) the pseudodimer model, both invoked to explain receptor triggering per se, and (3) two-step coreceptor recruitment to partially triggered TCRs leading to signal amplification. More recently it has been suggested that, in addition to initiating or augmenting TCR signaling, coreceptors effect antigen discrimination. But how can any of this be reconciled with TCR signaling occurring in the absence of CD4 or CD8, and with their interactions with pMHC being among the weakest specific protein-protein interactions ever described? Here, we review each theory of coreceptor function in light of the latest structural, biochemical, and functional data. We conclude that the oldest ideas are probably still the best, i.e., that their weak binding to MHC proteins and efficient association with Lck allow coreceptors to amplify weak incipient triggering of the TCR, without comprising TCR specificity.
Collapse
Affiliation(s)
- Alexander M. Mørch
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Štefan Bálint
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Simon J. Davis
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Michael L. Dustin
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
22
|
Thelen F, Witherden DA. Get in Touch With Dendritic Epithelial T Cells! Front Immunol 2020; 11:1656. [PMID: 32849572 PMCID: PMC7403176 DOI: 10.3389/fimmu.2020.01656] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
Innate and adaptive immune systems continuously interchange information and orchestrate their immune responses to protect the host. γδT cells play crucial roles, as they incorporate both innate and adaptive immune characteristics. Dendritic epidermal T cells (DETC) are specialized γδT cells, which are uniquely positioned to rapidly respond to skin wounds and infections. Their elongated dendrite morphology allows them to be in continuous contact with multiple neighboring keratinocytes and Langerhans cells. Cellular interactions are fundamental to the formation, activation and maintenance of immune cell functions during steady state and pathology. Recent technological advances, especially in the field of cellular imaging, have contributed greatly to the characterization of complex cellular interactions in a spatiotemporally resolved manner. In this review, we will highlight the often-underappreciated function of DETC and other γδT cells during steady state and an ongoing immune response. More specifically, we discuss how DETC-precursors are shaped in the fetal thymus during embryogenesis as well as how direct cell-cell interactions of DETC with neighboring epidermal cells shape skin homeostasis and effector functions. Furthermore, we will discuss seminal work and recent discoveries made in the γδT cell field, which have highlighted the importance of γδT cells in the skin, both in humans and mice.
Collapse
Affiliation(s)
- Flavian Thelen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| | - Deborah A Witherden
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
23
|
Ben Khalaf N, Al-Mashoor W, Saeed A, Al-Mehatab D, Taha S, Bakhiet M, Fathallah MD. The mouse intron-nested gene, Israa, is expressed in the lymphoid organs and involved in T-cell activation and signaling. Mol Immunol 2019; 111:209-219. [PMID: 31096062 DOI: 10.1016/j.molimm.2019.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 03/25/2019] [Accepted: 04/13/2019] [Indexed: 10/26/2022]
Abstract
We have previously reported Israa, immune-system-released activating agent, as a novel gene nested in intron 8 of the mouse Zmiz1 gene. We have also shown that Israa encodes for a novel FYN-binding protein and might be involved in the regulation of T-cell activation. In this report, we demonstrate that Israa gene product regulates the expression of a pool of genes involved in T-cell activation and signaling. Real time PCR and GFP knock-in expression analysis showed that Israa is transcribed and expressed in the spleen mainly by CD3+CD8+ cells as well as in the thymus by CD3+ (DP and DN), CD4+SP and CD8+SP cells at different developmental stages. We also showed that Israa is downregulated in T-cells following activation of T-cell receptor. Using yeast two-hybrid analysis, we identified ELF1, a transcription factor involved in T-cell regulation, as an ISRAA-binding partner. Transcriptomic analysis of an EL4 cell line overexpressing ISRAA revealed differential expression of several genes involved in T-cell signaling, activation and development. Among these genes, Prkcb, Mib2, Fos, Ndfip2, Cxxc5, B2m, Gata3 and Cd247 were upregulated whereas Itk, Socs3, Tigit, Ifng, Il2ra and FoxJ1 were downregulated. Our findings support the existence in mouse of a novel FYN-related T-cell regulation pathway involving the product of an intron-nested gene.
Collapse
Affiliation(s)
- Noureddine Ben Khalaf
- Department of Life Sciences, Health Biotechnology Program, College of Graduates Studies, Arabian Gulf University, Manama, Bahrain
| | - Wedad Al-Mashoor
- Department of Life Sciences, Health Biotechnology Program, College of Graduates Studies, Arabian Gulf University, Manama, Bahrain
| | - Azhar Saeed
- University of Michigan Medical School, MI, USA
| | - Dalal Al-Mehatab
- Department of Life Sciences, Health Biotechnology Program, College of Graduates Studies, Arabian Gulf University, Manama, Bahrain
| | - Safa Taha
- Department of Molecular Medicine, Princess Al-Jawhara Center for Genetics and Inherited Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Bahrain
| | - Moiz Bakhiet
- Department of Molecular Medicine, Princess Al-Jawhara Center for Genetics and Inherited Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Bahrain
| | - M Dahmani Fathallah
- Department of Life Sciences, Health Biotechnology Program, College of Graduates Studies, Arabian Gulf University, Manama, Bahrain.
| |
Collapse
|
24
|
Abstract
T cells are central to the vertebrate immune system. Two distinct types of T cells, αβT and γδT cells, express different types of T cell antigen receptors (TCRs), αβTCR and γδTCR, respectively, that are composed of different sets of somatically rearranged TCR chains and CD3 subunits. γδT cells have recently attracted considerable attention due to their ability to produce abundant cytokines and versatile roles in host defense, tissue regeneration, inflammation, and autoimmune diseases. Both αβT and γδT cells develop in the thymus. Unlike the development of αβT cells, which depends on αβTCR-mediated positive and negative selection, the development of γδT cells, including the requirement of γδTCR, has been less well understood. αβT cells differentiate into effector cells in the peripheral tissues, whereas γδT cells acquire effector functions during their development in the thymus. In this review, we will discuss the current state of knowledge of the molecular mechanism of TCR signal transduction and its role in the thymic development of γδT cells, particularly highlighting a newly discovered mechanism that controls proinflammatory γδT cell development.
Collapse
|
25
|
Kumar Singh P, Kashyap A, Silakari O. Exploration of the therapeutic aspects of Lck: A kinase target in inflammatory mediated pathological conditions. Biomed Pharmacother 2018; 108:1565-1571. [PMID: 30372858 DOI: 10.1016/j.biopha.2018.10.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/01/2018] [Accepted: 10/02/2018] [Indexed: 10/28/2022] Open
Abstract
Lck, a non-receptor src family kinase, plays a vital role in various cellular processes such as cell cycle control, cell adhesion, motility, proliferation and differentiation. As a 56 KDa protein, Lck phosphorylates tyrosine residues of various proteins such as ZAP-70, ITK and protein kinase C. The structure of Lck is comprised of three domains, one SH3 in tandem with a SH2 domain at the amino terminal and the kinase domain at the carboxy terminal. Physiologically, Lck is involved in the development, function and differentiation of T-cells. Additionally, Lck regulates neurite outgrowth and maintains long-term synaptic plasticity in neurons. Given a major role of Lck in cytokine production and T cell signaling, alteration in expression and activity of Lck may result in various diseased conditions like cancer, asthma, diabetes, rheumatoid arthritis, psoriasis, inflammatory bowel diseases such as Crohn's disease and ulcerative colitis, atherosclerosis etc. This article provides evidence and information establishing Lck as one of the therapeutic targets in various inflammation mediated pathophysiological conditions.
Collapse
Affiliation(s)
- Pankaj Kumar Singh
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug research, Punjabi University, Patiala, Punjab, 147002, India
| | - Aanchal Kashyap
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug research, Punjabi University, Patiala, Punjab, 147002, India
| | - Om Silakari
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug research, Punjabi University, Patiala, Punjab, 147002, India.
| |
Collapse
|
26
|
Zhou J, Zhang Q, Henriquez JE, Crawford RB, Kaminski NE. Lymphocyte-Specific Protein Tyrosine Kinase (LCK) is Involved in the Aryl Hydrocarbon Receptor-Mediated Impairment of Immunoglobulin Secretion in Human Primary B Cells. Toxicol Sci 2018; 165:322-334. [PMID: 29860352 PMCID: PMC6659013 DOI: 10.1093/toxsci/kfy133] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a cytosolic ligand-activated transcription factor involved in xenobiotic sensing, cell cycle regulation, and cell development. In humans, the activation of AHR by 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), a high affinity AHR-ligand, impairs the secretion of immunoglobulin M (IgM) to suppress humoral immunity. However, the mechanisms bridging the activation of AHR and the impairment of IgM secretion by human primary B cells remain poorly understood. Recent transcriptomic analysis revealed upregulation of lymphocyte-specific protein tyrosine kinase (LCK) in AHR-activated human primary B cells. LCK is a well-characterized tyrosine kinase that phosphorylates critical signaling proteins involved in activation and cytokine production in T cells. Conversely, the role of LCK in human primary B cells is not well understood. In the current studies, we have verified the transcriptomic finding by detecting AHR-mediated upregulation of LCK protein in human primary B cells. We also confirmed the role of AHR in the upregulation of LCK by using a specific AHR antagonist, which abolished the AHR-mediated increase of LCK. Furthermore, we have confirmed the role of LCK in the AHR-mediated suppression of IgM by using LCK specific inhibitors, which restored the IgM secretion by human B cells in the presence of TCDD. Collectively, the current studies demonstrate a novel role of LCK in IgM response and provide new insights into the mechanism for AHR-mediated impairment of immunoglobulin secretion by human primary B cells.
Collapse
Affiliation(s)
- Jiajun Zhou
- Department of Microbiology & Molecular Genetics
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - Qiang Zhang
- Department of Environmental Health, Rollins School of Public Health, Emory University, Georgia 30322
| | - Joseph E Henriquez
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - Robert B Crawford
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - Norbert E Kaminski
- Department of Microbiology & Molecular Genetics
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan 48824
| |
Collapse
|
27
|
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: 380] [Impact Index Per Article: 47.5] [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
|
28
|
Muro R, Nitta T, Nakano K, Okamura T, Takayanagi H, Suzuki H. γδTCR recruits the Syk/PI3K axis to drive proinflammatory differentiation program. J Clin Invest 2017; 128:415-426. [PMID: 29202478 DOI: 10.1172/jci95837] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/31/2017] [Indexed: 12/14/2022] Open
Abstract
γδT cells produce inflammatory cytokines and have been implicated in the pathogenesis of cancer, infectious diseases, and autoimmunity. The T cell receptor (TCR) signal transduction that specifically regulates the development of IL-17-producing γδT (γδT17) cells largely remains unclear. Here, we showed that the receptor proximal tyrosine kinase Syk is essential for γδTCR signal transduction and development of γδT17 in the mouse thymus. Zap70, another tyrosine kinase essential for the development of αβT cells, failed to functionally substitute for Syk in the development of γδT17. Syk induced the activation of the PI3K/Akt pathway upon γδTCR stimulation. Mice deficient in PI3K signaling exhibited a complete loss of γδT17, without impaired development of IFN-γ-producing γδT cells. Moreover, γδT17-dependent skin inflammation was ameliorated in mice deficient in RhoH, an adaptor known to recruit Syk. Thus, we deciphered lineage-specific TCR signaling and identified the Syk/PI3K pathway as a critical determinant of proinflammatory γδT cell differentiation.
Collapse
Affiliation(s)
- Ryunosuke Muro
- Department of Immunology and Pathology, Research Institute, National Center for Global Health and Medicine, Chiba, Japan.,Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Nitta
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Tadashi Okamura
- Department of Laboratory Animal Medicine, and.,Section of Animal Models, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Harumi Suzuki
- Department of Immunology and Pathology, Research Institute, National Center for Global Health and Medicine, Chiba, Japan
| |
Collapse
|
29
|
Abstract
Major histocompatibility complex (MHC) restriction is a unique feature of T cell antigen recognition. Mature T cells respond to antigenic nonself peptides bound to self-MHC molecules, but a sizeable fraction of peripheral T cells can also respond to nonself peptide-MHC (pMHC) complexes in the context of transplantation. MHC specificity of the T cell receptor (TCR) repertoire is shaped during thymic development. Two hypotheses have been proposed to explain MHC specificity of T cells. It has been suggested that MHC specificity is an intrinsic feature of TCR structure, mediated by the germline-encoded regions of the TCR sequence. In support of this model, an estimated 15% to 30% of preselection TCR repertoire is estimated to be MHC-specific. Moreover, structural studies have shown some degree of conserved binding topology for TCR-peptide MHC complexes. However, there is also evidence that MHC restriction can be imposed on the TCR repertoire during thymic development, and it has been proposed that the interaction of the Lck kinase with CD4 or CD8 coreceptors is critical for generation of MHC specificity. This review will discuss recent work on assessment of the preselection of TCR repertoire, molecular evidence for the germline encoded TCR bias for MHC, and for the coreceptor sequestration model in the context of alloreactivity and transplantation.
Collapse
|
30
|
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: 50] [Impact Index Per Article: 6.3] [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
|
31
|
Abstract
Based on the histological features and outcome, the current WHO classification separates thymomas into A, AB, B1, B2 and B3 subtypes. It is hypothesized that the type A thymomas are derived from the thymic medulla while the type B thymomas are derived from the cortex. Due to occasional histological overlap between the tumor subtypes creating difficulties in their separation, the aim of this study was to provide their proteomic characterization and identify potential immunohistochemical markers aiding in tissue diagnosis. Pair-wise comparison of neoplastic and normal thymus by liquid chromatography tandem mass spectrometry (LC-MS/MS) of formalin fixed paraffin embedded tissue revealed 61 proteins differentially expressed in thymomas compared to normal tissue. Hierarchical clustering showed distinct segregation of subtypes AB, B1 and B2 from that of A and B3. Most notably, desmoyokin, a protein that is encoded by the AHNAK gene, was associated with type A thymomas and medulla of normal thymus, by LC-MS/MS and immunohistochemistry. In this global proteomic characterization of the thymoma, several proteins unique to different thymic compartments and thymoma subtypes were identified. Among differentially expressed proteins, desmoyokin is a marker specific for thymic medulla and is potentially promising immunohistochemical marker in separation of type A and B3 thymomas.
Collapse
|
32
|
Fu G, Yu M, Chen Y, Zheng Y, Zhu W, Newman DK, Wang D, Wen R. Phospholipase Cγ1 is required for pre-TCR signal transduction and pre-T cell development. Eur J Immunol 2016; 47:74-83. [PMID: 27759161 DOI: 10.1002/eji.201646522] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/14/2016] [Accepted: 10/17/2016] [Indexed: 12/23/2022]
Abstract
Pre-T cell receptor (TCR) signaling is required for pre-T cell survival, proliferation, and differentiation from the CD4 and CD8 double negative (DN) to the double positive (DP) stage. However, the pre-TCR signal transduction pathway is not fully understood and the signaling molecules involved have not been completely identified. Phospholipase Cγ (PLCγ) 1 is an important signaling molecule that generates two second messengers, diacylglycerol and inositol 1,4,5-trisphosphate, that are important to mediate PKC activation and intracellular Ca2+ flux in many signaling pathways. Previously, we have shown that PLCγ1 is important for TCR-mediated signaling, development and T-cell activation, but the role of PLCγ1 in pre-TCR signal transduction and pre-T cell development is not known. In this study, we demonstrated that PLCγ1 expression level in pre-T cells was comparable to that in mature T cells. Deletion of PLCγ1 prior to the pre-TCR signaling stage partially blocked the DN3 to DN4 transition and reduced thymic cellularity. We also demonstrated that deletion of PLCγ1 impaired pre-T cell proliferation without affecting cell survival. Further study showed that deficiency of PLCγ1 impaired pre-TCR mediated Ca2+ flux and Erk activation. Thus our studies demonstrate that PLCγ1 is important for pre-TCR mediated signal transduction and pre-T cell development.
Collapse
Affiliation(s)
- Guoping Fu
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Mei Yu
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Yuhong Chen
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Yongwei Zheng
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Wen Zhu
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA.,Interdisciplinary Program in Biomedical Science, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Debra K Newman
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Demin Wang
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA.,Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Renren Wen
- The Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
33
|
Adam AP, Lowery AM, Martino N, Alsaffar H, Vincent PA. Src Family Kinases Modulate the Loss of Endothelial Barrier Function in Response to TNF-α: Crosstalk with p38 Signaling. PLoS One 2016; 11:e0161975. [PMID: 27603666 PMCID: PMC5014308 DOI: 10.1371/journal.pone.0161975] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/15/2016] [Indexed: 01/23/2023] Open
Abstract
Activation of Src Family Kinase (SFK) signaling is required for the increase in endothelial permeability induced by a variety of cytokines and growth factors. However, we previously demonstrated that activation of endogenous SFKs by expression of dominant negative C-terminal Src Kinase (DN-Csk) is not sufficient to decrease endothelial adherens junction integrity. Basal SFK activity has been observed in normal venular endothelia and was not associated with increased basal permeability. The basal SFK activity however was found to contribute to increased sensitivity of the venular endothelium to inflammatory mediator-induced leakage. How SFK activation achieves this is still not well understood. Here, we show that SFK activation renders human dermal microvascular endothelial cells susceptible to low doses of TNF-α. Treatment of DN-Csk-expressing cells with 50 pg/ml TNF-α induced a loss of TEER as well as drastic changes in the actin cytoskeleton and focal adhesion proteins. This synergistic effect was independent of ROCK or NF-κB activity. TNF-α-induced p38 signaling was required for the synergistic effect on barrier function, and activation of the p38 MAPK alone was also able to induce changes in permeability only in monolayers with active SFKs. These results suggest that the activation of endogenous levels of SFK renders the endothelial barrier more susceptible to low, physiologic doses of TNF-α through activation of p38 which leads to a loss of endothelial tight junctions.
Collapse
Affiliation(s)
- Alejandro P. Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States of America
- Department of Ophthalmology, Albany Medical College, Albany, New York, United States of America
- * E-mail: (PAV); (APA)
| | - Anthony M. Lowery
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States of America
| | - Nina Martino
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States of America
| | - Hiba Alsaffar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States of America
| | - Peter A. Vincent
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States of America
- * E-mail: (PAV); (APA)
| |
Collapse
|
34
|
Chiang YJ, Hodes RJ. T-cell development is regulated by the coordinated function of proximal and distal Lck promoters active at different developmental stages. Eur J Immunol 2016; 46:2401-2408. [PMID: 27469439 DOI: 10.1002/eji.201646440] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 07/05/2016] [Accepted: 07/26/2016] [Indexed: 12/21/2022]
Abstract
Expression of Lck, a T-cell lineage-specific tyrosine kinase critical for T-cell development and activation, can be mediated by either proximal or distal lck promoter. We generated BAC transgenic mice in which BAC lck promoter was deleted and bred these transgenes to an Lck knockout background. Lck-PROX mice, in which only the proximal promoter is functional, have maximal Lck protein and normal thymic development through CD4- CD8- double negative (DN) and CD4+ CD8+ double positive (DP) stages, but undetectable Lck later in development and reduced mature single positive thymocytes. In contrast, Lck-DIST mice, in which only distal promoter was functional, are deficient in Lck protein in DN and DP thymocytes and severely defective in early T-cell development, with a block at the DN3-DN4 beta checkpoint equivalent to complete Lck knockouts. The ability of the proximal lck promoter to support thymic development is independent of Fyn; while, in contrast, the distal lck promoter alone is completely unable to support development in the absence of Fyn. Notably, normal thymocyte development is restored by presence of both proximal and distal promoters, even when independently expressed on different lck genes. These results define distinct and complementary requirements for proximal and distal lck promoters during T-cell development.
Collapse
Affiliation(s)
- Y Jeffrey Chiang
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Richard J Hodes
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD, USA. .,National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
35
|
Rampoldi F, Bonrouhi M, Boehm ME, Lehmann WD, Popovic ZV, Kaden S, Federico G, Brunk F, Gröne HJ, Porubsky S. Immunosuppression and Aberrant T Cell Development in the Absence of N-Myristoylation. THE JOURNAL OF IMMUNOLOGY 2015; 195:4228-43. [DOI: 10.4049/jimmunol.1500622] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 09/01/2015] [Indexed: 01/01/2023]
|
36
|
Kovács M, Németh T, Jakus Z, Sitaru C, Simon E, Futosi K, Botz B, Helyes Z, Lowell CA, Mócsai A. The Src family kinases Hck, Fgr, and Lyn are critical for the generation of the in vivo inflammatory environment without a direct role in leukocyte recruitment. ACTA ACUST UNITED AC 2014; 211:1993-2011. [PMID: 25225462 PMCID: PMC4172222 DOI: 10.1084/jem.20132496] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Kovács et al. examine the role of the Src family kinases Hck, Fgr, and Lyn in immune cell–mediated inflammation. Using arthritis and skin inflammation models, the authors show that mice lacking hematopoietic Hck, Fgr, and Lyn are protected from these inflammatory diseases, showing loss of myeloid cell recruitment and lack of inflammatory mediator production. Unexpectedly, the three kinases are dispensable for the intrinsic migratory ability of myeloid cells. These finding may have clinical implications in rheumatic and skin diseases. Although Src family kinases participate in leukocyte function in vitro, such as integrin signal transduction, their role in inflammation in vivo is poorly understood. We show that Src family kinases play a critical role in myeloid cell–mediated in vivo inflammatory reactions. Mice lacking the Src family kinases Hck, Fgr, and Lyn in the hematopoietic compartment were completely protected from autoantibody-induced arthritis and skin blistering disease, as well as from the reverse passive Arthus reaction, with functional overlap between the three kinases. Though the overall phenotype resembled the leukocyte recruitment defect observed in β2 integrin–deficient (CD18−/−) mice, Hck−/−Fgr−/−Lyn−/− neutrophils and monocytes/macrophages had no cell-autonomous in vivo or in vitro migration defect. Instead, Src family kinases were required for the generation of the inflammatory environment in vivo and for the release of proinflammatory mediators from neutrophils and macrophages in vitro, likely due to their role in Fcγ receptor signal transduction. Our results suggest that infiltrating myeloid cells release proinflammatory chemokine, cytokine, and lipid mediators that attract further neutrophils and monocytes from the circulation in a CD18-dependent manner. Src family kinases are required for the generation of the inflammatory environment but not for the intrinsic migratory ability of myeloid cells.
Collapse
Affiliation(s)
- Miklós Kovács
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary MTA-SE "Lendület" Inflammation Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, and MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, 1094 Budapest, Hungary
| | - Tamás Németh
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary MTA-SE "Lendület" Inflammation Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, and MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, 1094 Budapest, Hungary
| | - Zoltán Jakus
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary MTA-SE "Lendület" Inflammation Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, and MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, 1094 Budapest, Hungary
| | - Cassian Sitaru
- Department of Dermatology, University Hospital Freiburg and BIOSS Centre for Biological Signalling Studies, 79104 Freiburg, Germany
| | - Edina Simon
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary MTA-SE "Lendület" Inflammation Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, and MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, 1094 Budapest, Hungary
| | - Krisztina Futosi
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary
| | - Bálint Botz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, and János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, and János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, and János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, and János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
| | - Clifford A Lowell
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143
| | - Attila Mócsai
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary MTA-SE "Lendület" Inflammation Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, and MTA-SE "Lendület" Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, 1094 Budapest, Hungary
| |
Collapse
|
37
|
Abstract
The functions of Natural Killer (NK) cells are regulated by a highly redundant set of germline-encoded surface receptors that can inhibit or activate NK cell activities. NK cells can be activated by cytokines or through the interaction with transformed or infected cells. This typically results in the production of cytokines, chemokines, and the induction of cellular cytotoxicity. However, the reactivity of NK cells is modulated on various levels and shaped by processes such as development, education, priming, exposure to antigens and cytokines, and the formation of memory-like phenotypes. Here, I will summarize our current understanding of these processes and describe how they influence NK cell reactivity on a molecular level.
Collapse
Affiliation(s)
- Carsten Watzl
- Leibniz Research Center for Working Environment and Human Factors, IfADo, Dortmund, Germany.
| |
Collapse
|
38
|
Requirement and redundancy of the Src family kinases Fyn and Lyn in perforin-dependent killing of Cryptococcus neoformans by NK cells. Infect Immun 2013; 81:3912-22. [PMID: 23918783 DOI: 10.1128/iai.00533-13] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Natural killer (NK) cells directly recognize and kill fungi, such as the pathogenic fungus Cryptococcus neoformans, via cytolytic mechanisms. However, the precise signaling pathways governing this NK cell microbicidal activity and the implications for fungal recognition are still unknown. Previously, it was reported that NK cell anticryptococcal activity is mediated through a conserved phosphatidylinositol 3-kinase-extracellular signal-regulated kinase 1/2 (PI3K-ERK1/2) pathway. Using YT (a human NK-like cell line) and primary human NK cells, we sought to identify the upstream, receptor-proximal signaling elements that led to fungal cytolysis. We demonstrate that Src family kinases were activated in response to C. neoformans. Furthermore, pharmacologic inhibition with an Src kinase inhibitor blocked C. neoformans-induced downstream activation of PI3K and ERK1/2 and abrogated cryptococcal killing. At the same time, the inhibitor disrupted the polarization of perforin-containing granules toward the NK cell-cryptococcal synapse but had no effect on conjugate formation between the organism and the NK cell. Finally, small interfering RNA (siRNA) double (but not single) knockdown of two Src family kinases, Fyn and Lyn, blocked cryptococcal killing. Together these data demonstrate a mechanism whereby the Src family kinases, Fyn and Lyn, redundantly mediate anticryptococcal activity through the activation of PI3K and ERK1/2, which in turn facilitates killing by inducing the polarization of perforin-containing granules to the NK cell-cryptococcal synapse.
Collapse
|
39
|
Xiong J, Parker BL, Dalheimer SL, Yankee TM. Interleukin-7 supports survival of T-cell receptor-β-expressing CD4(-) CD8(-) double-negative thymocytes. Immunology 2013; 138:382-91. [PMID: 23215679 DOI: 10.1111/imm.12050] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 11/30/2012] [Accepted: 12/05/2012] [Indexed: 01/17/2023] Open
Abstract
Among the milestones that occur during T-cell development in the thymus is the expression of T-cell receptor-β (TCR-β) and the formation of the pre-TCR complex. Signals emanating from the pre-TCR trigger survival, proliferation and differentiation of T-cell precursors. Although the pre-TCR is essential for these cell outcomes, other receptors, such as Notch and CXCR4, also contribute. Whether interleukin-7 (IL-7) participates in promoting the survival or proliferation of pre-TCR-expressing cells is controversial. We used in vitro and in vivo models of T-cell development to examine the function of IL-7 in TCR-β-expressing thymocytes. Culturing TCR-β-expressing CD4(-) CD8(-) double-negative thymocytes in an in vitro model of T-cell development revealed that IL-7 reduced the frequency of CD4(+) CD8(+) double-positive thymocytes at the time of harvest. The mechanism for this change in the percentage of double-positive cells was that IL-7 promoted the survival of thymocytes that had not yet differentiated. By preserving the double-negative population, IL-7 reduced the frequency of double-positive thymocytes. Interleukin-7 was not required for proliferation in the in vitro system. To follow this observation, we examined mice lacking CD127 (IL-7Rα). In addition to the known effect of CD127 deficiency on T-cell development before TCR-β expression, CD127 deficiency also impaired the development of TCR-β-expressing double-negative thymocytes. Specifically, we found that Bcl-2 expression and cell cycle progression were reduced in TCR-β-expressing double-negative thymocytes in mice lacking CD127. We conclude that IL-7 continues to function after TCR-β is expressed by promoting the survival of TCR-β-expressing double-negative thymocytes.
Collapse
Affiliation(s)
- Juan Xiong
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | | | |
Collapse
|
40
|
Ohsugi T. A transgenic mouse model of human T cell leukemia virus type 1-associated diseases. Front Microbiol 2013; 4:49. [PMID: 23483782 PMCID: PMC3592262 DOI: 10.3389/fmicb.2013.00049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 02/21/2013] [Indexed: 01/10/2023] Open
Abstract
Human T cell leukemia virus type 1 (HTLV-1) is the etiological agent of adult T cell leukemia/lymphoma (ATLL) and several inflammatory diseases. Tax, the protein encoded by HTLV-1, may be responsible for the development of the diseases caused by this virus. To investigate the pathogenic role of Tax, several transgenic mouse strains expressing Tax have been developed in recent years. These mice develop various tumors including large granular lymphocytic leukemia, as well as inflammatory diseases such as arthritis. These results suggest that Tax expression alone is sufficient to cause both malignant neoplastic diseases and inflammatory diseases. However, until recently, there were no tax transgenic mice that develop T cell leukemia and lymphoma resembling ATLL. The first successful induction of leukemia in T cells was pre-T cell leukemia generated in transgenic mice in which a mouse lymphocyte-specific protein tyrosine kinase p56lck (lck)-proximal promoter was used to express the tax gene in immature T cells. Subsequently, transgenic mice were established in which the lck-distal promoter was used to express Tax in mature T cells; these mice developed mature T cell leukemia and lymphoma that more closely resembled ATLL than did earlier mouse models.
Collapse
Affiliation(s)
- Takeo Ohsugi
- Division of Microbiology and Genetics, Institute of Resource Development and Analysis, Kumamoto University Kumamoto, Japan
| |
Collapse
|
41
|
Covello JM, Bird S, Morrison RN, Bridle AR, Battaglene SC, Secombes CJ, Nowak BF. Isolation of RAG-1 and IgM transcripts from the striped trumpeter (Latris lineata), and their expression as markers for development of the adaptive immune response. FISH & SHELLFISH IMMUNOLOGY 2013; 34:778-788. [PMID: 23291253 DOI: 10.1016/j.fsi.2012.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 06/01/2023]
Abstract
A partial sequence of the recombination activating gene-1 (RAG-1) and several full length sequences of the immunoglobulin M (IgM) heavy chain mRNA were obtained from the striped trumpeter (Latris lineata). The RAG-1 fragment consisted of 205 aa and fell within the core region of the open reading frame. The complete IgM heavy chain sequences translated into peptides ranging between 581 and 591 aa. Both genes showed good homology to other vertebrate sequences. The expression of the two genes was assessed throughout the early developmental stages of striped trumpeter larvae (5-100 dph) and used as markers to follow the ontogeny of the adaptive immune response. Using RT-PCR, RAG-1 mRNA expression was detectable at 5 dph and remained so until 80 dph, before becoming undetectable at 100 dph. IgM expression was also detectable at 5 dph, and remained so throughout. These patterns of expression may suggest that the striped trumpeter possess mature B cells with surface IgM at 100 dph. However, complete immunological competence is likely not reached until some time later. The early detection of IgM mRNA at 5 dph led to the investigation of its presence in oocytes. Both RAG-1 and IgM mRNA transcripts were detected in unfertilized oocytes, suggesting that they are maternally transferred. The biological significance of such a phenomenon remains to be investigated.
Collapse
Affiliation(s)
- J M Covello
- National Centre for Marine Conservation and Resource Sustainability, University of Tasmania, Private Bag 1370, Launceston, Tasmania 7250, Australia.
| | | | | | | | | | | | | |
Collapse
|
42
|
Kuhns MS, Badgandi HB. Piecing together the family portrait of TCR-CD3 complexes. Immunol Rev 2013; 250:120-43. [PMID: 23046126 DOI: 10.1111/imr.12000] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The pre-T-cell receptor (TCR)-, αβTCR-, and γδTCR-CD3 complexes are members of a family of modular biosensors that are responsible for driving T-cell development, activation, and effector functions. They inform essential checkpoint decisions by relaying key information from their ligand-binding modules (TCRs) to their signaling modules (CD3γε + CD3δε and CD3ζζ) and on to the intracellular signaling apparatus. Their actions shape the T-cell repertoire, as well as T-cell-mediated immunity; yet, the mechanisms that underlie their activity remain an enigma. As with any molecular machine, understanding how they function depends upon understanding how their parts fit and work together. In the 30 years since the initial biochemical and genetic characterizations of the αβTCR, the structure and function of the individual components of these family members have been extensively characterized. Cumulatively, this information has allowed us to piece together a portrait of the αβTCR-CD3 complex and outline the form of the remaining family members. Here we review the known structural and functional characteristics of the components of these TCR-CD3 complex family members. We then discuss how these data have informed our understanding of the architecture of the αβTCR-CD3 complex as well as their implications for the other family members. The intent is to provide a framework for considering: (i) how these thematically similar complexes diverge to execute their specific functions and (ii) how our knowledge of the form and function of these distinct family members can cross-inform our understanding of the other family members.
Collapse
Affiliation(s)
- Michael S Kuhns
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, USA.
| | | |
Collapse
|
43
|
Mukherjee S, Zhu J, Zikherman J, Parameswaran R, Kadlecek TA, Wang Q, Au-Yeung B, Ploegh H, Kuriyan J, Das J, Weiss A. Monovalent and multivalent ligation of the B cell receptor exhibit differential dependence upon Syk and Src family kinases. Sci Signal 2013; 6:ra1. [PMID: 23281368 DOI: 10.1126/scisignal.2003220] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Src and Syk families of kinases are two distinct sets of kinases that play critical roles in initiating membrane-proximal B cell receptor (BCR) signaling. However, unlike in other lymphocytes, such as T cells, the "division of labor" between Src family kinases (SFKs) and Syk in B cells is not well separated because both Syk and SFKs can phosphorylate immunoreceptor tyrosine-based activation motifs (ITAMs) present in proteins comprising the BCR. To understand why B cells require both SFKs and Syk for activation, we investigated the roles of both families of kinases in BCR signaling with computational modeling and in vitro experiments. Our computational model suggested that positive feedback enabled Syk to substantially compensate for the absence of SFKs when spatial clustering of BCRs was induced by multimeric ligands. We confirmed this prediction experimentally. In contrast, when B cells were stimulated by monomeric ligands that failed to produce BCR clustering, both Syk and SFKs were required for complete and rapid BCR activation. Our data suggest that SFKs could play a pivotal role in increasing BCR sensitivity to monomeric antigens of pathogens and in mediating a rapid response to soluble multimeric antigens of pathogens that can induce spatial BCR clustering.
Collapse
Affiliation(s)
- Sayak Mukherjee
- Battelle Center for Mathematical Medicine, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kwon HJ, Kim HS. Signaling for synergistic activation of natural killer cells. Immune Netw 2012; 12:240-6. [PMID: 23396805 PMCID: PMC3566418 DOI: 10.4110/in.2012.12.6.240] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 11/12/2012] [Accepted: 11/15/2012] [Indexed: 12/30/2022] Open
Abstract
Natural killer (NK) cells play a pivotal role in early surveillance against virus infection and cellular transformation, and are also implicated in the control of inflammatory response through their effector functions of direct lysis of target cells and cytokine secretion. NK cell activation toward target cell is determined by the net balance of signals transmitted from diverse activating and inhibitory receptors. A distinct feature of NK cell activation is that stimulation of resting NK cells with single activating receptor on its own cannot mount natural cytotoxicity. Instead, specific pairs of co-activation receptors are required to unleash NK cell activation via synergy-dependent mechanism. Because each co-activation receptor uses distinct signaling modules, NK cell synergy relies on the integration of such disparate signals. This explains why the study of the mechanism underlying NK cell synergy is important and necessary. Recent studies revealed that NK cell synergy depends on the integration of complementary signals converged at a critical checkpoint element but not on simple amplification of the individual signaling to overcome intrinsic activation threshold. This review focuses on the signaling events during NK cells activation and recent advances in the study of NK cell synergy.
Collapse
Affiliation(s)
- Hyung-Joon Kwon
- Department of Medicine, Graduate School, University of Ulsan College of Medicine, Seoul 138-736, Korea
| | | |
Collapse
|
45
|
Hauck F, Randriamampita C, Martin E, Gerart S, Lambert N, Lim A, Soulier J, Maciorowski Z, Touzot F, Moshous D, Quartier P, Heritier S, Blanche S, Rieux-Laucat F, Brousse N, Callebaut I, Veillette A, Hivroz C, Fischer A, Latour S, Picard C. Primary T-cell immunodeficiency with immunodysregulation caused by autosomal recessive LCK deficiency. J Allergy Clin Immunol 2012; 130:1144-1152.e11. [PMID: 22985903 DOI: 10.1016/j.jaci.2012.07.029] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 07/19/2012] [Accepted: 07/19/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND Signals emanating from the antigen T-cell receptor (TCR) are required for T-cell development and function. The T lymphocyte-specific protein tyrosine kinase (Lck) is a key component of the TCR signaling machinery. On the basis of its function, we considered LCK a candidate gene in patients with combined immunodeficiency. OBJECTIVE We identify and describe a child with a T-cell immunodeficiency caused by a homozygous missense mutation of the LCK gene (c.1022T>C) resulting from uniparental disomy. METHODS Genetic, molecular, and functional analyses were performed to characterize the Lck deficiency, and the associated clinical and immunologic phenotypes are reported. RESULTS The mutant LCK protein (p.L341P) was weakly expressed with no kinase activity and failed to reconstitute TCR signaling in LCK-deficient T cells. The patient presented with recurrent respiratory tract infections together with predominant early-onset inflammatory and autoimmune manifestations. The patient displayed CD4(+) T-cell lymphopenia and low levels of CD4 and CD8 expression on the T-cell surface. The residual T lymphocytes had an oligoclonal T-cell repertoire and exhibited a profound TCR signaling defect, with only weak tyrosine phosphorylation signals and no Ca(2+) mobilization in response to TCR stimulation. CONCLUSION We report a new form of T-cell immunodeficiency caused by a LCK gene defect, highlighting the essential role of Lck in human T-cell development and responses. Our results also point out that defects in the TCR signaling cascade often result in abnormal T-cell differentiation and functions, leading to an important risk factor for inflammation and autoimmunity.
Collapse
Affiliation(s)
- Fabian Hauck
- INSERM 768, Laboratoire du Développement Normal et Pathologique du Système Immunitaire, Hôpital Necker-Enfants Malades, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
The development of T cell in the thymus and the activation of mature T cells in the secondary lymphoid tissues require T cell to make adaptive responses to signaling molecules of environment. The activation of T cell receptor (TCR) signaling pathway could be induced by the interaction of the TCR and its co-receptor CD4 and CD8 with MHC/peptide complex. This process involves co-stimulatory molecules and signals mediated by cytokine receptors, which eventually leads to the occurrence of T cell immune response. The Src-family kinases lymphocyte-specific protein tyrosine kinase (Lck) and proto-oncogene tyrosine-protein kinase (Fyn) are expressed in T cells and serve as the signaling molecules that are activated downstream of TCR. These signaling molecules play key roles in development, positive selection, and peripheral maintenance of naive T cells and lymphopenia-induced proliferation of peripheral T cells. Both Lck and Fyn are required for each of these TCR-based signaling pathways, and Lck seems to be the major contributor, while Fyn can only supplement some functions of Lck. In this review, we discussed the mechanisms by which these two proteins perform functions in T cell development based on our current understanding.
Collapse
|
47
|
Kannan Y, Wilson MS. TEC and MAPK Kinase Signalling Pathways in T helper (T H) cell Development, T H2 Differentiation and Allergic Asthma. JOURNAL OF CLINICAL & CELLULAR IMMUNOLOGY 2012; Suppl 12:11. [PMID: 24116341 PMCID: PMC3792371 DOI: 10.4172/2155-9899.s12-011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Significant advances in our understanding of the signalling events during T cell development and differentiation have been made in the past few decades. It is clear that ligation of the T cell receptor (TCR) triggers a series of proximal signalling cascades regulated by an array of protein kinases. These orchestrated and highly regulated series of events, with differential requirements of particular kinases, highlight the disparities between αβ+CD4+ T cells. Throughout this review we summarise both new and old studies, highlighting the role of Tec and MAPK in T cell development and differentiation with particular focus on T helper 2 (TH2) cells. Finally, as the allergy epidemic continues, we feature the role played by TH2 cells in the development of allergy and provide a brief update on promising kinase inhibitors that have been tested in vitro, in pre-clinical disease models in vivo and into clinical studies.
Collapse
Affiliation(s)
- Yashaswini Kannan
- Division of Molecular Immunology, National Institute for Medical Research, MRC, London, NW7 1AA, UK
| | - Mark S. Wilson
- Division of Molecular Immunology, National Institute for Medical Research, MRC, London, NW7 1AA, UK
| |
Collapse
|
48
|
Gascoigne NRJ, Casas J, Brzostek J, Rybakin V. Initiation of TCR phosphorylation and signal transduction. Front Immunol 2011; 2:72. [PMID: 22566861 PMCID: PMC3342367 DOI: 10.3389/fimmu.2011.00072] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 11/21/2011] [Indexed: 01/14/2023] Open
Abstract
Recent data with CD8+ T cells show that the initial phase of T cell receptor (TCR) binding to MHC–peptide (MHCp) is quickly followed by a second, stronger, binding phase representing the binding of CD8 to the MHCp. This second phase requires signaling by a Src-family kinase such as Lck. These data point out two aspects of the initial stage of TCR signaling that have not yet been clearly resolved. Firstly, how and by which Src-family kinase, is the initial phosphorylation of CD3ζ accomplished, given that the Lck associated with the co-receptors (CD4 or CD8) is not yet available. Secondly, what is the mechanism by which the co-receptor is brought close to the bound TCR before the co-receptor binds to MHCp?
Collapse
Affiliation(s)
- Nicholas R J Gascoigne
- Department of Immunology and Microbial Science, The Scripps Research Institute La Jolla, CA, USA.
| | | | | | | |
Collapse
|
49
|
Activation or tolerance of natural killer cells is modulated by ligand quality in a nonmonotonic manner. Biophys J 2011; 99:2028-37. [PMID: 20923636 DOI: 10.1016/j.bpj.2010.07.061] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 07/19/2010] [Accepted: 07/22/2010] [Indexed: 11/21/2022] Open
Abstract
Natural killer (NK) cells extend important immune resistance in vertebrates by lysing infected and tumor cells. A fine balance between opposing signals generated by a diverse set of stimulatory and inhibitory NK-cell receptors determines the fate of target cells interacting with the NK cells. We have developed a mathematical model involving membrane proximal initial signaling events that provides novel mechanistic insights into how activation of NK cells is modulated by the half-life of receptor-ligand interaction and ligand concentrations. We show that strong stimulatory ligands produce digital activation, whereas weaker stimulatory ligands can mediate inhibition by strengthening the signals generated by inhibitory ligands, as indicated in experiments in knockout mice. We find under certain conditions, counterintuitively, inhibitory receptors can help mediate activation instead of inhibition. Mechanistic insights gained from NK-cell signaling can facilitate understanding of complex signaling responses that occur due to cross talk between dueling signaling pathways in other cell types.
Collapse
|
50
|
Campbell M, Lie WR, Zhao J, Hayes D, Mistry J, Kung HJ, Luciw PA, Khan IH. Multiplex analysis of Src family kinase signaling by microbead suspension arrays. Assay Drug Dev Technol 2010; 8:488-96. [PMID: 20482378 DOI: 10.1089/adt.2009.0255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
There is renewed interest in the Src family of protein tyrosine kinases (SFKs) as a result of their potential utility as molecular targets for cancer therapy. This protein family consists of 9 nonreceptor tyrosine kinases that, although implicated in a diverse array of cellular functions, possess a similar modular structure. Here we describe a simple and efficient multiplex microbead immunoassay (MMIA), based on Luminex xMAP technology, which allows for the simultaneous detection of 8 phosphorylated SFKs in a single assay. Microbead sets identifiable by unique fluorescence were individually coated with antibodies specific for an individual SFK member. Detection of phosphorylated SFKs was accomplished using a secondary antibody directed against phosphotyrosine. The assay requires < or = 10 microg of cell lysate or nanogram amounts of purified SFK. The use of a generic secondary antibody allows for the expansion of the assay to include any other tyrosine kinase for which a specific antibody exists. Using either mammalian cell lines or purified, recombinant kinases as the SFK source, we demonstrate the utility of the assay by evaluating the phosphorylation status of SFK members following several in vitro manipulations designed to modulate the phosphotyrosine content of the kinases. These results show that the SFK multiplex assay is a robust tool to investigate the function of SFKs in basic and potentially in clinical research.
Collapse
Affiliation(s)
- Mel Campbell
- Center for Comparative Medicine, University of California, Davis, California 95616, USA
| | | | | | | | | | | | | | | |
Collapse
|