1
|
Gad AZ, Morris JS, Godret-Miertschin L, Montalvo MJ, Kerr SS, Berger H, Lee JCH, Saadeldin AM, Abu-Arja MH, Xu S, Vasileiou S, Brock RM, Fousek K, Sheha MF, Srinivasan M, Li Y, Saeedi A, R. Levental K, Leen AM, Mamonkin M, Carisey A, Varadarajan N, Hegde M, Joseph SK, Levental I, Mukherjee M, Ahmed N. Molecular dynamics at immune synapse lipid rafts influence the cytolytic behavior of CAR T cells. SCIENCE ADVANCES 2025; 11:eadq8114. [PMID: 39792660 PMCID: PMC11721525 DOI: 10.1126/sciadv.adq8114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025]
Abstract
Chimeric antigen receptor T cells (CART) targeting CD19 through CD28.ζ signaling induce rapid lysis of leukemic blasts, contrasting with persistent tumor control exhibited by 4-1BB.ζ-CART. We reasoned that molecular dynamics at the CART immune synapse (CARIS) could explain differences in their tumor rejection kinetics. We observed that CD28.ζ-CART engaged in brief highly lethal CARIS and mastered serial killing, whereas 4-1BB.ζ-CART formed lengthy CARIS and relied on robust expansion and cooperative killing. We analyzed CARIS membrane lipid rafts (mLRs) and found that, upon tumor engagement, CD28.ζ-CAR molecules rapidly but transiently translocated into mLRs, mobilizing the microtubular organizing center and lytic granules to the CARIS. This enabled fast CART recovery and sensitivity to low target site density. In contrast, gradual accumulation of 4-1BB.ζ-CAR and LFA-1 molecules at mLRs built mechanically tonic CARIS mediating chronic Fas ligand-based killing. The differences in CD28.ζ- and 4-1BB.ζ-CARIS dynamics explain the distinct cytolytic behavior of CART and can guide engineering of more adaptive effective cellular products.
Collapse
Affiliation(s)
- Ahmed Z. Gad
- Interdepartmental Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jessica S. Morris
- Interdepartmental Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lea Godret-Miertschin
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Melisa J. Montalvo
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, USA
| | - Sybrina S. Kerr
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Immunology & Microbiology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Harrison Berger
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jessica C. H. Lee
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Amr M. Saadeldin
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mohammad H. Abu-Arja
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shuo Xu
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Spyridoula Vasileiou
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rebecca M. Brock
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kristen Fousek
- Interdepartmental Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mohamed F. Sheha
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Madhuwanti Srinivasan
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yongshuai Li
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, USA
| | - Arash Saeedi
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, USA
| | - Kandice R. Levental
- Department of Molecular Physiology and Biological Physics, Center for Molecular and Cell Physiology, University of Virginia, Charlottesville, VA 22903, USA
| | - Ann M. Leen
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Maksim Mamonkin
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexandre Carisey
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Navin Varadarajan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, USA
| | - Meenakshi Hegde
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sujith K. Joseph
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, Center for Molecular and Cell Physiology, University of Virginia, Charlottesville, VA 22903, USA
| | - Malini Mukherjee
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital William T. Shearer Center for Human Immunobiology, Houston, TX 77030, USA
| | - Nabil Ahmed
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
2
|
Kasten-Jolly J, Lawrence DA. Perfluorooctanesulfonate (PFOS) and perfluorooctanoic acid (PFOA) modify in vitro mitogen- and antigen-induced human peripheral blood mononuclear cell (PBMC) responses. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2022; 85:715-737. [PMID: 35611390 DOI: 10.1080/15287394.2022.2075816] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Environmental contaminants perfluorooctanoate (PFOA) and perfluorooctanesulfonate (PFOS) are present in human serum at the highest concentration among all per- and polyfluoroalkyl substances (PFAS). Serum concentrations as high as 500 ng and 3000 ng PFOA/ml have been detected in individuals living near contamination sites and those occupationally exposed, respectively. Animal and human studies indicated that PFOA and PFOS at these serum concentrations perturb the immune system. The aim of this study was to examine the effects of in vitro exposure of human peripheral blood mononuclear cells (PBMC) to 1, 10, or 100 µM PFOA or PFOS in a medium with serum (RPMI-1640 + 5% human AB serum) on the measurement of proliferation, T cell activation, generation of memory T cells, and cytokine production/secretion. In addition, these immune system parameters were assessed for PBMC in a serum-free medium (OpSFM), which was stimulated with phytohemagglutinin (PHA) (2.5 µg/ml) or influenza vaccine antigen (0.625 µg/ml Flu Ag). PFOS decreased proliferation stimulated by PHA or Flu Ag. With Flu Ag stimulation, PFOA and PFOS inhibited the generation of memory T cells in a concentration-dependent manner. In OpSFM, PFOA and PFOS produced no marked change in proliferation and no inhibition of T cell activation. Cytokines measured in the media with Luminex methodology indicated decreased PBMC secretion of IFN-γ by PFOA and PFOS in medium with serum, but no alteration in OpSFM. The results indicated that changes in immune parameters due to PFOA or PFOS following Flu Ag stimulation are medium (±serum) dependent.
Collapse
Affiliation(s)
| | - David A Lawrence
- Department of Health, Wadsworth Center, Albany, NY, USA
- School of Public Health, University at Albany, Rensselaer, NY, USA
| |
Collapse
|
3
|
Inokuchi JI, Nagafuku M. Gangliosides in T cell development and function of mice. Glycoconj J 2022; 39:229-238. [DOI: 10.1007/s10719-021-10037-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/19/2021] [Accepted: 12/29/2021] [Indexed: 11/30/2022]
|
4
|
Liu L, Limsakul P, Meng X, Huang Y, Harrison RES, Huang TS, Shi Y, Yu Y, Charupanit K, Zhong S, Lu S, Zhang J, Chien S, Sun J, Wang Y. Integration of FRET and sequencing to engineer kinase biosensors from mammalian cell libraries. Nat Commun 2021; 12:5031. [PMID: 34413312 PMCID: PMC8376904 DOI: 10.1038/s41467-021-25323-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 07/30/2021] [Indexed: 01/01/2023] Open
Abstract
The limited sensitivity of Förster Resonance Energy Transfer (FRET) biosensors hinders their broader applications. Here, we develop an approach integrating high-throughput FRET sorting and next-generation sequencing (FRET-Seq) to identify sensitive biosensors with varying substrate sequences from large-scale libraries directly in mammalian cells, utilizing the design of self-activating FRET (saFRET) biosensor. The resulting biosensors of Fyn and ZAP70 kinases exhibit enhanced performance and enable the dynamic imaging of T-cell activation mediated by T cell receptor (TCR) or chimeric antigen receptor (CAR), revealing a highly organized ZAP70 subcellular activity pattern upon TCR but not CAR engagement. The ZAP70 biosensor elucidates the role of immunoreceptor tyrosine-based activation motif (ITAM) in affecting ZAP70 activation to regulate CAR functions. A saFRET biosensor-based high-throughput drug screening (saFRET-HTDS) assay further enables the identification of an FDA-approved cancer drug, Sunitinib, that can be repurposed to inhibit ZAP70 activity and autoimmune-disease-related T-cell activation.
Collapse
Affiliation(s)
- Longwei Liu
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, CA, USA
| | - Praopim Limsakul
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, CA, USA
- Center of Excellence for Trace Analysis and Biosensor, Division of Physical Science, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
| | - Xianhui Meng
- Department of Cell Biology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, P.R. China
| | - Yan Huang
- Department of Chemistry and Chemical Engineering, Hunan University, Changsha, P.R. China
| | - Reed E S Harrison
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, CA, USA
| | - Tse-Shun Huang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, CA, USA
- BioLegend, San Diego, CA, USA
| | - Yiwen Shi
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, CA, USA
| | - Yiyan Yu
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, CA, USA
| | - Krit Charupanit
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Sheng Zhong
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, CA, USA
| | - Shaoying Lu
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, CA, USA
| | - Jin Zhang
- Department of Pharmacology, University of California, San Diego, CA, USA
| | - Shu Chien
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, CA, USA
- Department of Medicine, University of California, San Diego, CA, USA
| | - Jie Sun
- Department of Cell Biology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, P.R. China.
| | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, CA, USA.
| |
Collapse
|
5
|
Kim MS, Lee A, Cho D, Kim TS. AIMP1 regulates TCR signaling and induces differentiation of regulatory T cells by interfering with lipid raft association. Biochem Biophys Res Commun 2019; 514:875-880. [PMID: 31084930 DOI: 10.1016/j.bbrc.2019.05.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 05/05/2019] [Indexed: 02/07/2023]
Abstract
In addition to a role in translation, AIMP1 is secreted to affect various immune cells, such as macrophages, dendritic cells, B cells, and natural killer cells. However, the direct effects of AIMP1 on T cells have not yet been reported. In this study, we investigated whether AIMP1 could modulate T cell responses directly. Results revealed that AIMP1 significantly inhibited T cell receptor (TCR)-dependent activation and proliferation of CD4 T cells, as well as decreased TCR stimuli-induced Ca2+ influx in CD4 T cells. In addition, microscopic analysis revealed that lipid raft association in response to TCR engagement was significantly reduced in the presence of AIMP1, and the phosphorylation of PLCγ and PI3K was also down-regulated in CD4 T cells by AIMP1. Furthermore, AIMP1 specifically enhanced the differentiation of regulatory T (Treg) cells, while it had no effect on T helper type 1 (Th1), type 2 (Th2), and type 17 (Th17) cell differentiation. Collectively, these results indicate that AIMP1 affects T cells directly by down-regulating TCR signaling complex formation and inducing Treg cell differentiation in CD4 T cells.
Collapse
MESH Headings
- Animals
- Calcium/immunology
- Calcium/metabolism
- Cell Differentiation/drug effects
- Cytokines/genetics
- Cytokines/immunology
- Cytokines/pharmacology
- Female
- Gene Expression Regulation
- Immunophenotyping
- Ion Transport/drug effects
- Lymphocyte Activation/drug effects
- Membrane Microdomains/drug effects
- Membrane Microdomains/immunology
- Membrane Microdomains/metabolism
- Mice
- Mice, Inbred C57BL
- Phosphatidylinositol 3-Kinase/genetics
- Phosphatidylinositol 3-Kinase/immunology
- Phospholipase C gamma/genetics
- Phospholipase C gamma/immunology
- Phosphorylation/drug effects
- Primary Cell Culture
- Receptors, Antigen, T-Cell/antagonists & inhibitors
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes, Helper-Inducer/cytology
- T-Lymphocytes, Helper-Inducer/drug effects
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Myun Soo Kim
- Institute of Convergence Science, Korea University, 5-ga, Anam-dong, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Arim Lee
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, 5-ga, Anam-dong, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Daeho Cho
- Institute of Convergence Science, Korea University, 5-ga, Anam-dong, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Tae Sung Kim
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, 5-ga, Anam-dong, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| |
Collapse
|
6
|
Inokuchi JI, Inamori KI, Kabayama K, Nagafuku M, Uemura S, Go S, Suzuki A, Ohno I, Kanoh H, Shishido F. Biology of GM3 Ganglioside. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 156:151-195. [PMID: 29747813 DOI: 10.1016/bs.pmbts.2017.10.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Since the successful molecular cloning in 1998 of GM3 synthase (GM3S, ST3GAL5), the enzyme responsible for initiating biosynthesis of all complex gangliosides, the efforts of our research group have been focused on clarifying the physiological and pathological implications of gangliosides, particularly GM3. We have identified isoforms of GM3S proteins having distinctive lengths of N-terminal cytoplasmic tails, and found that these cytoplasmic tails define subcellular localization, stability, and in vivo activity of GM3S isoforms. Our studies of the molecular pathogenesis of type 2 diabetes, focused on interaction between insulin receptor and GM3 in membrane microdomains, led to a novel concept: type 2 diabetes and certain other lifestyle-related diseases are membrane microdomain disorders resulting from aberrant expression of gangliosides. This concept has enhanced our understanding of the pathophysiological roles of GM3 and related gangliosides in various diseases involving chronic inflammation, such as insulin resistance, leptin resistance, and T-cell function and immune disorders (e.g., allergic asthma). We also demonstrated an essential role of GM3 in murine and human auditory systems; a common pathological feature of GM3S deficiency is deafness. This is the first direct link reported between gangliosides and auditory functions.
Collapse
Affiliation(s)
- Jin-Ichi Inokuchi
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
| | - Kei-Ichiro Inamori
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | | | - Masakazu Nagafuku
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Satoshi Uemura
- Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Shinji Go
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Akemi Suzuki
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Isao Ohno
- Center for Medical Education, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Hirotaka Kanoh
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Fumi Shishido
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| |
Collapse
|
7
|
Schatzlmaier P, Supper V, Göschl L, Zwirzitz A, Eckerstorfer P, Ellmeier W, Huppa JB, Stockinger H. Rapid multiplex analysis of lipid raft components with single-cell resolution. Sci Signal 2015; 8:rs11. [PMID: 26396269 DOI: 10.1126/scisignal.aac5584] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Lipid rafts, a distinct class of highly dynamic cell membrane microdomains, are integral to cell homeostasis, differentiation, and signaling. However, their quantitative examination is challenging when working with rare cells, developmentally heterogeneous cell populations, or molecules that only associate weakly with lipid rafts. We present a fast biochemical method, which is based on lipid raft components associating with the nucleus upon partial lysis during centrifugation through nonionic detergent. Requiring little starting material or effort, our protocol enabled the multidimensional flow cytometric quantitation of raft-resident proteins with single-cell resolution, thereby assessing the membrane components from a few cells in complex cell populations, as well as their dynamics resulting from cell signaling, differentiation, or genetic mutation.
Collapse
Affiliation(s)
- Philipp Schatzlmaier
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
| | - Verena Supper
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
| | - Lisa Göschl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria. Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria
| | - Alexander Zwirzitz
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
| | - Paul Eckerstorfer
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
| | - Wilfried Ellmeier
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
| | - Johannes B Huppa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria.
| | - Hannes Stockinger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria.
| |
Collapse
|
8
|
Abstract
Cancer cells have long been known to fuel their pathogenic growth habits by sustaining a high glycolytic flux, first described almost 90 years ago as the so-called Warburg effect. Immune cells utilize a similar strategy to generate the energy carriers and metabolic intermediates they need to produce biomass and inflammatory mediators. Resting lymphocytes generate energy through oxidative phosphorylation and breakdown of fatty acids, and upon activation rapidly switch to aerobic glycolysis and low tricarboxylic acid flux. T cells in patients with rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) have a disease-specific metabolic signature that may explain, at least in part, why they are dysfunctional. RA T cells are characterized by low adenosine triphosphate and lactate levels and increased availability of the cellular reductant NADPH. This anti-Warburg effect results from insufficient activity of the glycolytic enzyme phosphofructokinase and differentiates the metabolic status in RA T cells from those in cancer cells. Excess production of reactive oxygen species and a defect in lipid metabolism characterizes metabolic conditions in SLE T cells. Owing to increased production of the glycosphingolipids lactosylceramide, globotriaosylceramide and monosialotetrahexosylganglioside, SLE T cells change membrane raft formation and fail to phosphorylate pERK, yet hyperproliferate. Borrowing from cancer metabolomics, the metabolic modifications occurring in autoimmune disease are probably heterogeneous and context dependent. Variations of glucose, amino acid and lipid metabolism in different disease states may provide opportunities to develop biomarkers and exploit metabolic pathways as therapeutic targets.
Collapse
Affiliation(s)
- Zhen Yang
- Department of Medicine, Stanford University School of Medicine, CCSR Building Rm 2225, 269 Campus Drive West, Stanford, CA, 94305-5166, USA.
| | - Eric L Matteson
- Division of Rheumatology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA.
| | - Jörg J Goronzy
- Department of Medicine, Stanford University School of Medicine, CCSR Building Rm 2225, 269 Campus Drive West, Stanford, CA, 94305-5166, USA.
| | - Cornelia M Weyand
- Department of Medicine, Stanford University School of Medicine, CCSR Building Rm 2225, 269 Campus Drive West, Stanford, CA, 94305-5166, USA.
| |
Collapse
|
9
|
Inokuchi JI, Nagafuku M, Ohno I, Suzuki A. Distinct selectivity of gangliosides required for CD4⁺ T and CD8⁺ T cell activation. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:98-106. [PMID: 25193136 DOI: 10.1016/j.bbalip.2014.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 06/03/2014] [Accepted: 07/13/2014] [Indexed: 01/25/2023]
Abstract
T cells compose a crucial part of the immune system and require activation. The first step of T cell activation is triggered by the movement of one of their surface molecules, known as T cell receptor, into localized regions of cell membrane known as lipid rafts. Molecules called gangliosides are known to be major components of lipid rafts, but their role in T-cell activation remains to be elucidated. This review summarizes recent findings that different types of T cells require distinct ganglioside types for the activation. Control of ganglioside expression would offer a strategy targeting for specific T-cell subpopulations to treat immune diseases. This article is part of a Special Issue entitled Linking transcription to physiology in lipodomics.
Collapse
Affiliation(s)
- Jin-Ichi Inokuchi
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai 981-8558, Japan.
| | - Masakazu Nagafuku
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai 981-8558, Japan
| | - Isao Ohno
- Department of Pathophysiology, Tohoku Pharmaceutical University, Sendai 981-8558, Japan
| | - Akemi Suzuki
- Institute of Glycoscience, Tokai University, Hiratsuka, Kanagawa 259-1292, Japan
| |
Collapse
|
10
|
Maciolek JA, Pasternak JA, Wilson HL. Metabolism of activated T lymphocytes. Curr Opin Immunol 2014; 27:60-74. [PMID: 24556090 DOI: 10.1016/j.coi.2014.01.006] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/20/2013] [Accepted: 01/14/2014] [Indexed: 01/04/2023]
Abstract
Activated T cells undergo metabolic reprogramming which promotes glycolytic flux and lactate production as well as elevated production of lipids, proteins, nucleic acids and other carbohydrates (i.e. induction of biomass) even in the presence of oxygen. Activated T cells show induced expression of, among other things, Glucose Transporter 1 and several glycolytic enzymes, including ADP-Dependent Glucokinase and the low affinity isoform Pyruvate Kinase-M2 (which promote glycolytic flux), as well Glutamine Transporters and Glycerol-3-phosphate Dehydrogenase 2 which make available glutamate and glycerol-3-phosphate as mitochondrial energy sources. Intracellular leucine concentrations critically regulate mammalian target of rapamycin (mTOR) signaling to promote Th1, Th2, and Th17 CD4(+) T effector cell differentiation. In contrast, T regulatory (Treg) cells are generated when AMP-Activating Protein Kinase signaling is activated and mTOR activation is suppressed. Unlike effector CD4(+) and CD8(+) T cells, Tregs and memory T cells oxidize fatty acids for fuel. Effector and memory T cells perform different functions and thus show distinct metabolic profiles which are exquisitely controlled by cellular signaling. Upon activation, T cells express the insulin and leptin receptors on their surface and become sensitive to insulin signaling and nutrient availability and show changes in differentiation. Thus, metabolism and nutrient availability influence T cell activation and function.
Collapse
Affiliation(s)
- Jason A Maciolek
- Vaccine and Infectious Disease Organization (VIDO)-Home of the International Vaccine Centre (InterVac), University of Saskatchewan, 120 Veterinary Road, Saskatoon, S7N 5E3, Canada
| | - J Alex Pasternak
- Vaccine and Infectious Disease Organization (VIDO)-Home of the International Vaccine Centre (InterVac), University of Saskatchewan, 120 Veterinary Road, Saskatoon, S7N 5E3, Canada
| | - Heather L Wilson
- Vaccine and Infectious Disease Organization (VIDO)-Home of the International Vaccine Centre (InterVac), University of Saskatchewan, 120 Veterinary Road, Saskatoon, S7N 5E3, Canada.
| |
Collapse
|
11
|
Mancini RJ, Tom JK, Esser-Kahn AP. Covalently Coupled Immunostimulant Heterodimers. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201306551] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
12
|
Mancini RJ, Tom JK, Esser-Kahn AP. Covalently coupled immunostimulant heterodimers. Angew Chem Int Ed Engl 2013; 53:189-92. [PMID: 24259411 DOI: 10.1002/anie.201306551] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 10/16/2013] [Indexed: 02/02/2023]
Abstract
We report increased stimulation of dendritic cells via heterodimers of immunostimulants formed at a discrete molecular distance. Many vaccines present spatially organized agonists to immune cell receptors. These receptors cluster suggesting that signaling is increased by spatial organization and receptor proximity, but this has not been directly tested for multiple, unique receptors. In this study we probe the spatial aspect of immune cell activation using heterodimers of two covalently attached immunostimulants.
Collapse
Affiliation(s)
- Rock J Mancini
- Department of Chemistry, University of California, Irvine, 3038A Frederick Reines Hall, Irvine, CA 92697-2025 (USA)
| | | | | |
Collapse
|
13
|
Inokuchi JI, Nagafuku M, Ohno I, Suzuki A. Heterogeneity of gangliosides among T cell subsets. Cell Mol Life Sci 2013; 70:3067-75. [PMID: 23233133 PMCID: PMC11114073 DOI: 10.1007/s00018-012-1208-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Revised: 10/07/2012] [Accepted: 11/05/2012] [Indexed: 01/16/2023]
Abstract
Gangliosides are major components of highly organized membrane microdomains or rafts, yet little is known about the role of gangliosides in raft organization. This is also the case of gangliosides in TCR-mediated activation. Comprehensive structural analysis of gangliosides in the primary thymocytes and CD4(+) T and CD8(+) T cells was not achieved due to technical difficulties. We have found that CD8(+) T cells express very high levels of o-series gangliosides, but on the other hand, CD4(+) T cells preferably express a-series gangliosides. In the TCR-dependent activation, CD4(+) T cells selectively require a-series gangliosides, but CD8(+) T cells do require only o-series gangliosides but not a-series gangliosides. Ganglioside GM3 synthase-deficient mice lacking a-series gangliosides neither exhibited the TCR-dependent activation of CD4(+) T nor developed ovalbumin-induced allergic airway inflammation. These findings imply that the distinct expression pattern of ganglioside species in CD4(+) and CD8(+) T cells define the immune function of each T cell subset.
Collapse
Affiliation(s)
- Jin-ichi Inokuchi
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, 981-8558, Japan.
| | | | | | | |
Collapse
|
14
|
Fulop T, Le Page A, Garneau H, Azimi N, Baehl S, Dupuis G, Pawelec G, Larbi A. Aging, immunosenescence and membrane rafts: the lipid connection. LONGEVITY & HEALTHSPAN 2012; 1:6. [PMID: 24764511 PMCID: PMC3886260 DOI: 10.1186/2046-2395-1-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 07/13/2012] [Indexed: 11/10/2022]
Abstract
The decreased efficiency of immune responses in older people is partly a consequence of alterations in T lymphocyte functions caused by modifications in the early events of signal transduction. Several alterations in the signaling pathways of T lymphocytes have been described in older humans and animals. A unifying cause could be modifications in the physicochemical properties of the plasma membrane resulting from changes in its lipid composition and the distribution and function of lipid rafts (LR). The latter serve to assemble the initial components of the signaling cascade. Accumulating data suggest that the function of plasma membrane LR is altered with aging; we hypothesize that this would significantly contribute to immune dysregulation. The role of aging and cholesterol in LR functions in T lymphocytes is reviewed and discussed here.
Collapse
Affiliation(s)
- Tamas Fulop
- Department of Medicine, Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 12th Avenue North, Sherbrooke, Qc, J1H 5N4, Canada ; Research Center on Aging, University of Sherbrooke, 1036, rue Belvedere Sud, Sherbrooke, Qc, J1H 4C4, Canada
| | - Aurélie Le Page
- Department of Medicine, Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 12th Avenue North, Sherbrooke, Qc, J1H 5N4, Canada
| | - Hugo Garneau
- Department of Medicine, Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 12th Avenue North, Sherbrooke, Qc, J1H 5N4, Canada
| | - Naheed Azimi
- Department of Medicine, Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 12th Avenue North, Sherbrooke, Qc, J1H 5N4, Canada
| | - Sarra Baehl
- Department of Medicine, Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 12th Avenue North, Sherbrooke, Qc, J1H 5N4, Canada
| | - Gilles Dupuis
- Department of Biochemistry, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 12th Avenue North, Sherbrooke, Qc, J1H 5N4, Canada
| | - Graham Pawelec
- Center for Medical Research, Tübingen Aging and Tumor Immunology Group, University of Tübingen, Waldhörnlestrasse 22, Tübingen, D-72072, Germany
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Immunos Building/Biopolis, Agency for Science Technology and Research (ASTAR), 8A Biomedical Grove, Singapore, 138648, Singapore
| |
Collapse
|
15
|
CD4 and CD8 T cells require different membrane gangliosides for activation. Proc Natl Acad Sci U S A 2012; 109:E336-42. [PMID: 22308377 DOI: 10.1073/pnas.1114965109] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Initial events of T-cell activation involve movement of the T-cell receptor into lipid rafts. Gangliosides are major components of lipid rafts. While investigating T-cell activation in ganglioside-deficient mice, we observed that CD4(+) and CD8(+) T cells required different ganglioside subsets for activation. Activation of CD4(+) T cells from GM3 synthase-null mice, deficient in GM3-derived gangliosides, is severely compromised, whereas CD8(+) T-cell activation is normal. Conversely, in cells from GM2/GD2 synthase-null mice, expressing only GM3 and GD3, CD4(+) T-cell activation is normal, whereas CD8(+) T-cell activation is deficient. Supplementing the cells with the corresponding missing gangliosides restores normal activation. GM3 synthase-null mice do not develop experimental asthma. Distinct expression patterns of ganglioside species in CD4(+) T and CD8(+) T cells, perhaps in uniquely functional lipid rafts, define immune functions in each T-cell subset. Control of ganglioside expression would offer a strategy targeting for specific T-cell subpopulations to treat immune diseases.
Collapse
|
16
|
Irles C, Arias-Martinez J, Guzmán-Bárcenas J, Ortega A. Plasma membrane subdomain partitioning of Lck in primary human T lymphocytes. Can J Physiol Pharmacol 2010; 88:487-96. [PMID: 20555418 DOI: 10.1139/y09-125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Uncovering the plasma membrane distribution of tyrosine kinase Lck is crucial to understanding T lymphocyte triggering. Several studies of Lck species partitioning have given contradictory results. We decided to re-address this point by using phospho-specific antibodies to characterize active and inactive Lck partitioning in raft and non-raft membranes from primary human peripheral blood T lymphocytes. We show that most inactive Lck was localized in rafts and was associated with nearly all CD4 coreceptors and its negative regulator Csk in resting cells, while T cell receptor (TCR) engagement promoted a sustained dephosphorylation of inactive Lck. In contrast, active Lck had a more discrete distribution interacting with only a small number of CD4 coreceptors, and the kinase showed a rapid and short phosphorylation after TCR triggering. The differences in distribution and kinetics may be related to T lymphocyte signalling threshold modulation by Lck species and suggest how TCR triggering is first initiated. This study furthers our knowledge of the TCR activation model in primary human T lymphocytes.
Collapse
Affiliation(s)
- Claudine Irles
- Departamento de Bioquímica y Biología Molecular, Instituto Nacional de Perinatologia "Isidro Espinoza de los Reyes", México D.F, C.P. 11000, México
| | | | | | | |
Collapse
|
17
|
Increased expression of ganglioside GM1 in peripheral CD4+ T cells correlates soluble form of CD30 in Systemic Lupus Erythematosus patients. J Biomed Biotechnol 2010; 2010:569053. [PMID: 20625494 PMCID: PMC2896695 DOI: 10.1155/2010/569053] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Revised: 04/14/2010] [Accepted: 04/26/2010] [Indexed: 12/18/2022] Open
Abstract
Gangliosides GM1 is a good marker of membrane microdomains (lipid rafts) with important function in cellular activation processes. In this study we found that GM1 expression on CD4+ T cells and memory T cells (CD45RO/CD4) were dramatic increased after stimulation with phytohaemagglutinin in vitro. Next, we examined the GM1 expression on peripheral blood CD4+ T cells and CD8+ T cells from 44 patients with SLE and 28 healthy controls by flow cytometry. GM1 expression was further analyzed with serum soluble CD30 (sCD30), IL-10, TNF-alpha and clinical parameters. The mean fluorescence intensity of GM1 on CD4+ T cells from patients with SLE was significantly higher than those from healthy controls, but not on CD8+ T cells. Increased expression of GM1 was more marked on CD4+/CD45RO+ memory T cells from active SLE patients. Patients with SLE showed significantly elevated serum sCD30 and IL-10, but not TNF-alpha levels. In addition, we found that enhanced GM1 expression on CD4+ T cells from patients with SLE positively correlated with high serum levels of sCD30 and IgG as well as disease activity (SLEDAI scores). Our data suggested the potential role of aberrant lipid raft/GM1 on CD4+ T cells and sCD30 in the pathogenesis of SLE.
Collapse
|
18
|
|
19
|
Willemsen RA, Sebestyén Z, Ronteltap C, Berrevoets C, Drexhage J, Debets R. CD8 alpha coreceptor to improve TCR gene transfer to treat melanoma: down-regulation of tumor-specific production of IL-4, IL-5, and IL-10. THE JOURNAL OF IMMUNOLOGY 2006; 177:991-8. [PMID: 16818755 DOI: 10.4049/jimmunol.177.2.991] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Therapeutic success of TCR gene transfer to treat tumors depends on the ability of redirected T cells to become activated upon tumor recognition in vivo. Help provided by tumor-specific Th1 cells is reported to relieve T cells from an anergized state and to induce tumor regression. We recently demonstrated the ability to generate melanoma-specific Th1 cells by genetic introduction of both a CD8-dependent TCR and the CD8alpha coreceptor into CD4+ T cells. In this study, we analyzed a TCR that binds Ag independently of CD8, a property generally preferred to induce tumor-specific T cell responses, and addressed the contribution of CD8alpha following introduction into TCR-transduced CD4+ T cells. To this end, primary human CD4+ T cells were gene transferred with a high-avidity TCR, and were shown not only to bind peptide/MHC class I, but also to effectively kill Ag-positive tumor cells in the absence of CD8alpha. The introduction of CD8alpha up-regulates the tumor-specific production of TNF-alpha and IL-2 to some extent, but significantly down-regulates production of IL-4, IL-5, and IL-10 in CD4+ T cells. The introduction of a mutated cysteine motif in CD8alpha, which prevents its binding to LCK and linker for activation of T cells, did not adversely affect expression and T cell cytotoxicity, but counteracted the CD8alpha-mediated down-regulation of IL-4 and IL-5, but not IL-10. In conclusion, CD8alpha down-regulates the production of major Th2-type cytokines, in part mediated by LCK and/or linker for activation of T cells, and may induce differentiation of tumor-specific Th1 cells, which makes this coreceptor an interesting candidate to improve the clinical potential of TCR gene transfer to treat cancer.
Collapse
MESH Headings
- Amino Acid Motifs/genetics
- CD8 Antigens/genetics
- CD8 Antigens/physiology
- CD8 Antigens/therapeutic use
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Line
- Cell Line, Tumor
- Cysteine/genetics
- Cytotoxicity, Immunologic/genetics
- Down-Regulation/genetics
- Down-Regulation/immunology
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Gene Transfer Techniques
- HLA-A2 Antigen/immunology
- HLA-A2 Antigen/metabolism
- Humans
- Interleukin-10/antagonists & inhibitors
- Interleukin-10/biosynthesis
- Interleukin-4/antagonists & inhibitors
- Interleukin-4/biosynthesis
- Interleukin-5/antagonists & inhibitors
- Interleukin-5/biosynthesis
- Melanoma/genetics
- Melanoma/immunology
- Melanoma/therapy
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Protein Binding/genetics
- Protein Binding/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/therapeutic use
- Th1 Cells/cytology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Transduction, Genetic/methods
- gp100 Melanoma Antigen
Collapse
Affiliation(s)
- Ralph A Willemsen
- Laboratory of Tumor Immunology, Unit of Clinical and Tumor Immunology, Department of Medical Oncology, Erasmus Medisch Centrum (MC)-Daniel den Hoed Cancer Center, 3008 AE Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
20
|
Larbi A, Dupuis G, Khalil A, Douziech N, Fortin C, Fülöp T. Differential role of lipid rafts in the functions of CD4+ and CD8+ human T lymphocytes with aging. Cell Signal 2006; 18:1017-30. [PMID: 16236485 DOI: 10.1016/j.cellsig.2005.08.016] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2005] [Revised: 08/22/2005] [Accepted: 08/22/2005] [Indexed: 02/04/2023]
Abstract
Lipid rafts are critical to the assembly of the T-cell receptor (TCR) signaling machinery. It is not known whether lipid raft properties differ in CD4+ and CD8+ T cells and whether there are age-related differences that may account in part for immune senescence. Data presented here showed that time-dependent interleukin-2 (IL-2) production was different between CD4+ and CD8+ T cells. The defect in IL-2 production by CD4+ T cells was not due to lower levels of expression of the TCR or CD28. There was a direct correlation between the activation of p56(Lck) and LAT and their association/recruitment with the lipid raft fractions of CD4+ and CD8+ T cells. p56Lck, LAT and Akt/PKB were weakly phosphorylated in lipid rafts of stimulated CD4+ T cells of elderly as compared to young donors. Lipid rafts undergo changes in their lipid composition (ganglioside M1, cholesterol) in CD4+ and CD8+ T cells of elderly individuals. This study emphasizes the differential role of lipid rafts in CD4+ and CD8+ T-cell activation in aging and suggests that the differential localization of CD28 may explain disparities in response to stimulation in human aging.
Collapse
Affiliation(s)
- Anis Larbi
- Research Center on Aging, 1036 Belvedere Street South, Sherbrooke, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
21
|
Abstract
Functional polarization of leukocytes is a requisite to accomplish immune function. Immune synapse formation or chemotaxis requires asymmetric redistribution of membrane receptors, signaling molecules and the actin cytoskeleton. There is increasing evidence that compartmentalization of the plasma membrane into distinct lipid microdomains is pivotal in establishing and maintaining leukocyte polarity. Specific rafts assemble into large-scale domains to create plasma membrane asymmetries at specific cell locations, thus coordinating temporally and spatially cell signaling in these processes. In this review we discuss the roles of lipid rafts as organizers of T lymphocyte polarity during cell activation and migration.
Collapse
Affiliation(s)
- Santos Mañes
- Department of Immunology and Oncology, National Center of Biotechnology/Consejo Superior de Investigaciones Científicas, Madrid, Spain.
| | | |
Collapse
|
22
|
Kovacs B, Parry RV, Ma Z, Fan E, Shivers DK, Freiberg BA, Thomas AK, Rutherford R, Rumbley CA, Riley JL, Finkel TH. Ligation of CD28 by its natural ligand CD86 in the absence of TCR stimulation induces lipid raft polarization in human CD4 T cells. THE JOURNAL OF IMMUNOLOGY 2006; 175:7848-54. [PMID: 16339520 DOI: 10.4049/jimmunol.175.12.7848] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Stimulation of resting CD4 T cells with anti-CD3/CD28-coated beads leads to rapid polarization of lipid rafts (LRs). It has been postulated that a major role of costimulation is to facilitate LR aggregation. CD86 is up-regulated or expressed aberrantly on immune cells in a wide array of autoimmune and infectious diseases. Using an Ig fusion with the extracellular domain of CD86 (CD86Ig) bound to a magnetic bead or K562 cells expressing CD86, we demonstrated that ligation of CD28 by its natural ligand, but not by Ab, induced polarization of LRs at the cell-bead interface of fresh human CD4 T cells in the absence of TCR ligation. This correlated with activation of Vav-1, increase of the intracellular calcium concentration, and nuclear translocation of NF-kappaB p65, but did not result in T cell proliferation or cytokine production. These studies show, for the first time, that LR polarization can occur in the absence of TCR triggering, driven solely by the CD28/CD86 interaction. This result has implications for mechanisms of T cell activation. Abnormalities in this process may alter T and B cell tolerance and susceptibility to infection.
Collapse
Affiliation(s)
- Birgit Kovacs
- Division of Rheumatology, Children's Hospital of Philadelphia, and Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
The molecular events and the protein components that are involved in signalling by the T cell receptor (TCR) for antigen have been extensively studied. Activation of signalling cascades following TCR stimulation depends on the phosphorylation of the receptor by the tyrosine kinase Lck, which localizes to the cytoplasmic face of the plasma membrane by virtue of its post-translational modification. However, the precise order of events during TCR phosphorylation at the plasma membrane, remains to be defined. A current theory that describes early signalling events incorporates the function of lipid rafts, microdomains at the plasma membrane with distinct lipid and protein composition. Lipid rafts have been implicated in diverse biological functions in mammalian cells. In T cells, molecules with a key role in TCR signalling, including Lck, localize to these domains. Importantly, mutant versions of these proteins which fail to localise to raft domains were unable to support signalling by the TCR. Biochemical studies using purified detergent-resistant membranes (DRM) and confocal microscopy have suggested that upon stimulation, the TCR and Lck-containing lipid rafts may come into proximity allowing phosphorylation of the receptor. Further, there are data suggesting that phosphorylation of the TCR could depend on a transient increase in Lck activity that takes place within lipid rafts to initiate signalling. Current results and a model of how lipid rafts may regulate TCR signalling are discussed.
Collapse
Affiliation(s)
- Panagiotis S Kabouridis
- Bone & Joint Research Unit, William Harvey Research Institute, Queen Mary's School of Medicine & Dentistry, University of London, UK.
| |
Collapse
|
24
|
Ermini L, Secciani F, La Sala GB, Sabatini L, Fineschi D, Hale G, Rosati F. Different glycoforms of the human GPI-anchored antigen CD52 associate differently with lipid microdomains in leukocytes and sperm membranes. Biochem Biophys Res Commun 2005; 338:1275-83. [PMID: 16266689 DOI: 10.1016/j.bbrc.2005.10.082] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2005] [Accepted: 10/12/2005] [Indexed: 10/25/2022]
Abstract
CD52 is a human GPI-anchored antigen, expressed exclusively in the immune system and part of the reproductive system (epididymal cells). Sperm cells acquire the antigen from the epididymal secretions when transiting in the epididymal corpus and cauda. The peptide backbone of CD52, consisting of only 12 aminoacids, is generally considered no more than a scaffold for post-translational modifications, such as GPI-anchor and especially N-glycosylation which occur at the third asparagine. The latter modification is highly heterogeneous, especially in the reproductive system, giving rise to many different glycoforms, some of which are tissue specific. A peculiar O-glycan-containing glycoform is also found in reproductive and immune systems. We determined to locate CD52 in microdomains of leukocytes and sperm membranes using two antibodies: (1) CAMPATH-1G, the epitope of which includes the last three aminoacids and part of the GPI-anchor of glycoforms present in leukocytes and sperm cells; (2) anti-gp20, the epitope of which belongs to the unique O-glycan-bearing glycoform also present in both cell types. Using a Brij 98 solubilization protocol and sucrose gradient partition we demonstrated that the CD52 glycoforms recognized by both antibodies are markers of typical raft microdomains in leukocytes, whereas in capacitated sperm the O-glycoform is included in GM3-rich microdomains different from the cholesterol and GM1-rich lipid rafts with which CAMPATH antigen is stably associated. The importance of the association between GM3 and O-glycans for formation of specialized microdomains was confirmed by heterologous CD52 insertion experiments. When prostasomes from human seminal fluid were incubated with rat sperm from different epididymal regions, the CD52 glycoform recognized by anti-gp20 decorated rat epididymal corpus and cauda sperm, associated with the same low-cholesterol GM3-rich sperm membrane fractions as in human sperm. The glycoforms recognized by CAMPATH-1G were not found in rat sperm. The relationship between this differential insertion and differences in glycosylation of rat and human CD52 is discussed.
Collapse
Affiliation(s)
- L Ermini
- Department of Evolutionary Biology, University of Siena, Italy
| | | | | | | | | | | | | |
Collapse
|
25
|
Davanture S, Leignadier J, Milani P, Soubeyran P, Malissen B, Malissen M, Schmitt-Verhulst AM, Boyer C. Selective defect in antigen-induced TCR internalization at the immune synapse of CD8 T cells bearing the ZAP-70(Y292F) mutation. THE JOURNAL OF IMMUNOLOGY 2005; 175:3140-9. [PMID: 16116204 DOI: 10.4049/jimmunol.175.5.3140] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cbl proteins have been implicated in ligand-induced TCR/CD3 down-modulation, but underlying mechanisms are unclear. We analyzed the effect of mutation of a cbl-binding site on ZAP-70 (ZAP-Y292F) on dynamics, internalization, and degradation of the TCR/CD3 complex in response to distinct stimuli. Naive CD8 T cells expressing the P14 transgenic TCR from ZAP-Y292F mice were selectively affected in TCR/CD3 down-modulation in response to antigenic stimulation, whereas neither anti-CD3 Ab-, and PMA-induced TCR down-modulation, nor constitutive receptor endocytosis/cycling were impaired. We further established that the defect in TCR/CD3 down-modulation in response to Ag was paralleled by an impaired TCR/CD3 internalization and CD3zeta degradation. Analysis of T/APC conjugates revealed that delayed redistribution of TCR at the T/APC contact zone was paralleled by a delay in TCR internalization in the synaptic zone in ZAP-Y292F compared with ZAP-wild-type T cells. Cbl recruitment to the synapse was also retarded in ZAP-Y292F T cells, although F-actin and LFA-1 redistribution was similar for both cell types. This study identifies a step involving ZAP-70/cbl interaction that is critical for rapid internalization of the TCR/CD3 complex at the CD8 T cell/APC synapse.
Collapse
Affiliation(s)
- Suzel Davanture
- Centre d'Immunologie de Marseille-Luminy, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université de la Méditerranée, Marseille, Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Ise W, Nakamura K, Shimizu N, Goto H, Fujimoto K, Kaminogawa S, Hachimura S. Orally tolerized T cells can form conjugates with APCs but are defective in immunological synapse formation. THE JOURNAL OF IMMUNOLOGY 2005; 175:829-38. [PMID: 16002680 DOI: 10.4049/jimmunol.175.2.829] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Oral tolerance is systemic immune hyporesponsiveness induced by the oral administration of soluble Ags. Hyporesponsiveness of Ag-specific CD4 T cells is responsible for this phenomenon. However, the molecular mechanisms underlying the hyporesponsive state of these T cells are not fully understood. In the present study, we investigated the ability of orally tolerized T cells to form conjugates with Ag-bearing APCs and to translocate TCR, protein kinase C-theta (PKC-theta), and lipid rafts into the interface between T cells and APCs. Orally tolerized T cells were prepared from the spleens of OVA-fed DO11.10 mice. Interestingly, the orally tolerized T cells did not show any impairment in the formation of conjugates with APCs. The conjugates were formed in a LFA-1-dependent manner. Upon antigenic stimulation, the tolerized T cells could indeed activate Rap1, which is critical for LFA-1 activation and thus cell adhesion. However, orally tolerized T cells showed defects in the translocation of TCR, PKC-theta, and lipid rafts into the interface between T cells and APCs. Translocation of TCR and PKC-theta to lipid raft fractions upon antigenic stimulation was also impaired in the tolerized T cells. Ag-induced activation of Vav, Rac1, and cdc42, which are essential for immunological synapse and raft aggregation, were down-regulated in orally tolerized T cells. These results demonstrate that orally tolerized T cells can respond to specific Ags in terms of conjugate formation but not with appropriate immunological synapse formation. This may account for the hyporesponsive state of orally tolerized T cells.
Collapse
Affiliation(s)
- Wataru Ise
- Department of Applied Biological Chemistry, University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Hoshino N, Katayama N, Shibasaki T, Ohishi K, Nishioka J, Masuya M, Miyahara Y, Hayashida M, Shimomura D, Kato T, Nakatani K, Nishii K, Kuribayashi K, Nobori T, Shiku H. A novel role for Notch ligand Delta-1 as a regulator of human Langerhans cell development from blood monocytes. J Leukoc Biol 2005; 78:921-9. [PMID: 16037408 DOI: 10.1189/jlb.1204746] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Human Langerhans cells (LCs) are of hematopoietic origin, but cytokine regulation of their development is not fully understood. Notch ligand Delta-1 is expressed in a proportion of the skin. Granulocyte-macrophage colony-stimulating factor (GM-CSF) and transforming growth factor-beta1 (TGF-beta1) are also secreted in the skin. We report here that Delta-1, in concert with GM-CSF and TGF-beta1, induces the differentiation of human CD14(+) blood monocytes into cells that express LC markers: CD1a, Langerin, cutaneous lymphocyte-associated antigen, CC chemokine receptor 6, E-cadherin, and Birbeck granules. The resulting cells display phagocytic activity and chemotaxis to macrophage inflammatory protein-1alpha (MIP-1alpha). In response to CD40 ligand and tumor necrosis factor alpha, the cells acquire a mature phenotype of dendritic cells that is characterized by up-regulation of human leukocyte antigen (HLA)-ABC, HLA-DR, CD80, CD86, CD40, and CD54 and appearance of CD83. These cells in turn show chemotaxis toward MIP-1beta and elicit activation of CD8(+) T cells and T helper cell type 1 polarization of CD4(+) T cells. Thus, blood monocytes can give rise to LCs upon exposure to the skin cytokine environment consisting of Delta-1, GM-CSF, and TGF-beta1, which may be, in part, relevant to the development of human epidermal LCs. Our results extend the functional scope of Notch ligand delta-1 in human hematopoiesis.
Collapse
Affiliation(s)
- Natsuki Hoshino
- Department of Hematology and Oncology, Mie University School of Medicine, Mie 514-8507, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Memory T cells exhibit low activation thresholds and mediate rapid effector responses when recalled by antigen; contrasting the higher activation threshold, slower responses and predominant IL-2 production by naive T cells. While the sequence of intracellular events coupling the T cell-receptor (TCR) to naive T cell activation is well characterized, biochemical control of memory T cell differentiation and function remains undefined. In this review, we will discuss recent developments in T cell-receptor signal transduction as they pertain to memory T cells, and will discuss how signal dampening may drive memory generation, and more efficient spatial organization of signaling molecules may promote rapid recall responses.
Collapse
Affiliation(s)
- Meena R Chandok
- Division of Transplantation, Department of Surgery, University of Maryland School of Medicine, MSTF Building, Room 400, 685 W. Baltimore St., Baltimore, MD 21201, USA
| | | |
Collapse
|
29
|
Quintana A, Hoth M. Apparent cytosolic calcium gradients in T-lymphocytes due to fura-2 accumulation in mitochondria. Cell Calcium 2005; 36:99-109. [PMID: 15193858 DOI: 10.1016/j.ceca.2004.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2003] [Revised: 01/05/2004] [Accepted: 01/06/2004] [Indexed: 11/25/2022]
Abstract
Fura-2 is the most common dye to measure cytosolic Ca2+ concentrations ([Ca2+]i). To facilitate simultaneous imaging of many cells while preserving their cytosolic environment, fura-2 is often loaded into the cytosol in its membrane-permeant ester form. It has been reported that small amounts of fura-2 accumulate in intracellular compartments, an effect that is usually neglected. We show that either focal or non-focal stimulation methods induce large [Ca2+]i gradients in T-lymphocytes during both, Ca2+ release and Ca2+ influx across the plasma membrane. Interfering with mitochondrial Ca2+ homeostasis and by labeling mitochondria with MitoTracker, we demonstrate that [Ca2+]i gradients co-localize with mitochondria and are attributable to mitochondrial fura-2 sequestration. Gradients could not be avoided by different loading protocols, compromising measurements of "real" [Ca2+]i gradients following T-cell stimulation. They were observed in human blood and lamina propria lymphocytes, Jurkat T-cells, mast cells, but not to the same extent in HEK-293 cells. Finally, we show that T-lymphocytes can be efficiently loaded with the membrane-impermeant fura-2 salt by electroporation and by osmotic lysis of pinocytic vesicles, which result in the loss of [Ca2+]i gradients. These methods are therefore suitable to study localized Ca2+ signals in large populations of T-cells while preserving their cytosolic integrity.
Collapse
Affiliation(s)
- Ariel Quintana
- Department of Physiology, University of the Saarland, 66421 Hamburg/Saal, Germany.
| | | |
Collapse
|
30
|
Friedländer E, Arndt-Jovin DJ, Nagy P, Jovin TM, Szöllosi J, Vereb G. Signal transduction of erbB receptors in trastuzumab (Herceptin) sensitive and resistant cell lines: Local stimulation using magnetic microspheres as assessed by quantitative digital microscopy. Cytometry A 2005; 67:161-71. [PMID: 16163699 DOI: 10.1002/cyto.a.20173] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND ErbB2 (HER-2), a member of the epidermal growth factor (EGF) receptor family, is a class I transmembrane receptor tyrosine kinase. Although erbB2 has no known physiologic ligand, it can form complexes with other members of the family and undergo transactivation of its very potent kinase activity, thereby initiating downstream signaling and cell proliferation. ErbB2 is a frequent pathologic marker in ductal invasive breast carcinomas and is targeted by using a specific humanized monoclonal antibody, trastuzumab (Herceptin). The antibody is effective in only 20% to 50% of erbB2-positive tumors, and this resistance, as yet poorly understood, constitutes a major therapeutic challenge. METHODS Magnetic microspheres coated with ligands or antibodies are widely used for separation of proteins and cells and allow localized, high intensity, and precisely timed stimulation of cells. We used EGF- and trastuzumab-covered paramagnetic microspheres, quantitative confocal laser scanning microscopy, and digital image processing to investigate the (trans)activation of and local signal propagation from erbB1 and erbB2 on trastuzumab sensitive and resistant carcinoma cell lines expressing these receptors at high levels. RESULTS On A431 cells expressing high levels of endogenous erbB1 and transfected erbB2-mYFP (A4-erbB2-mYFP F4 cell line), EGF-coupled-microspheres activated erbB1 and transactivated erbB2-mYFP. In two other cell lines with comparable erbB2 expression but lower levels of erbB1, EGF microspheres transactivated erbB2 less efficiently. Trastuzumab in solution activated erbB2 on A4-erbB2-mYFP and the trastuzumab sensitive SKBR-3 cells, but only negligibly on the resistant JIMT-1 cells that showed a 10 times higher K(d) for the antibody. Nevertheless, pronounced erbB2 activation and tyrosine phosphorylation could be detected after stimulation with trastuzumab-coupled microspheres in all cell lines, although transactivation of erbB1 was negligible. Receptor phosphorylation was restricted to the immediate proximity of the microspheres, i.e., receptor clusters external to these locations remained inactive. CONCLUSION ErbB1 ligand and erbB2 specific antibody attached to magnetic microspheres are efficient tools in assessing erbB activation, localized signal propagation, and erbB heterodimer formation. Trastuzumab coupled to microspheres is more efficient at accessing erbB2 and activating it than trastuzumab in solution.
Collapse
Affiliation(s)
- Elza Friedländer
- Department of Biophysics and Cell Biology, Research Center for Molecular Medicine, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | | | | | | | | | | |
Collapse
|
31
|
Nguyen DH, Giri B, Collins G, Taub DD. Dynamic reorganization of chemokine receptors, cholesterol, lipid rafts, and adhesion molecules to sites of CD4 engagement. Exp Cell Res 2004; 304:559-69. [PMID: 15748900 DOI: 10.1016/j.yexcr.2004.11.022] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2004] [Revised: 11/16/2004] [Accepted: 11/18/2004] [Indexed: 11/18/2022]
Abstract
T cell polarization and redistribution of cellular components are critical to processes such as activation, migration, and potentially HIV infection. Here, we investigate the effects of CD4 engagement on the redistribution and localization of chemokine receptors, CXCR4 and CCR5, adhesion molecules, and lipid raft components including cholesterol, GM1, and glycosyl-phosphatidylinositol (GPI)-anchored proteins. We demonstrate that anti-CD4-coated beads (alpha CD4-B) rapidly induce co-capping of chemokine receptors as well as GPI-anchored proteins and adhesion molecules with membrane cholesterol and lipid rafts on human T cell lines and primary T cells to the area of bead-cell contact. This process was dependent on the presence of cellular cholesterol, cytoskeletal reorganization, and lck signaling. Lck-deficient JCaM 1.6 cells failed to cap CXCR4 or lipid rafts to alpha CD4-B. Biochemical analysis reveals that CXCR4 and LFA-1 are recruited to lipid rafts upon CD4 but not CD45 engagement. Furthermore, we also demonstrate T cell capping of both lipid rafts and chemokine receptors at sites of contact with HIV-infected cells, despite the binding of an HIV inhibitory mAb to CXCR4. We conclude that cell surface rearrangements in response to CD4 engagement may serve as a means to enhance cell-to-cell signaling at the immunological synapse and modulate chemokine responsiveness, as well as facilitate HIV entry and expansion by synaptic transmission.
Collapse
Affiliation(s)
- Dzung H Nguyen
- Laboratory of Immunology, National Institute on Aging, Intramural Research Program, NIH, DHHS, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA
| | | | | | | |
Collapse
|
32
|
Abstract
Lateral compartmentalization of the plasma membrane into domains is a key feature of immune cell activation and subsequent immune effector functions. Here, we will review the high diversity of membrane domains, ranging from elementary lipid rafts, envisioned as dynamic and small domains (in the tens of nm), to relatively stable microm-scale membrane domains, which form the immunologic synapse of T lymphocytes. We will discuss the relationship between these different types of plasma membrane domains and how raft lipid- and protein-controlled interactions and cell biological processes cooperate to generate functional domains that mediate lymphocyte activity.
Collapse
Affiliation(s)
- Thomas Harder
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | | |
Collapse
|
33
|
Razzaq TM, Ozegbe P, Jury EC, Sembi P, Blackwell NM, Kabouridis PS. Regulation of T-cell receptor signalling by membrane microdomains. Immunology 2004; 113:413-26. [PMID: 15554919 PMCID: PMC1782593 DOI: 10.1111/j.1365-2567.2004.01998.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2004] [Revised: 09/09/2004] [Accepted: 09/15/2004] [Indexed: 01/04/2023] Open
Abstract
There is now considerable evidence suggesting that the plasma membrane of mammalian cells is compartmentalized by functional lipid raft microdomains. These structures are assemblies of specialized lipids and proteins and have been implicated in diverse biological functions. Analysis of their protein content using proteomics and other methods revealed enrichment of signalling proteins, suggesting a role for these domains in intracellular signalling. In T lymphocytes, structure/function experiments and complementary pharmacological studies have shown that raft microdomains control the localization and function of proteins which are components of signalling pathways regulated by the T-cell antigen receptor (TCR). Based on these studies, a model for TCR phosphorylation in lipid rafts is presented. However, despite substantial progress in the field, critical questions remain. For example, it is unclear if membrane rafts represent a homogeneous population and if their structure is modified upon TCR stimulation. In the future, proteomics and the parallel development of complementary analytical methods will undoubtedly contribute in further delineating the role of lipid rafts in signal transduction mechanisms.
Collapse
Affiliation(s)
- Tahir M Razzaq
- Bone and Joint Research Unit, William Harvey Research Institute, Queen Mary's School of Medicine and Dentistry, Queen Mary's College, London
| | | | | | | | | | | |
Collapse
|
34
|
Maus MV, Kovacs B, Kwok WW, Nepom GT, Schlienger K, Riley JL, Allman D, Finkel TH, June CH. Extensive replicative capacity of human central memory T cells. THE JOURNAL OF IMMUNOLOGY 2004; 172:6675-83. [PMID: 15153483 DOI: 10.4049/jimmunol.172.11.6675] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To characterize the replicative capacity of human central memory (T(CM)) CD4 T cells, we have developed a defined culture system optimized for the ex vivo expansion of Ag-specific CD4(+) T cells. Artificial APCs (aAPCs) consisting of magnetic beads coated with Abs to HLA class II and a costimulatory Ab to CD28 were prepared; peptide-charged HLA class II tetramers were then loaded on the beads to provide Ag specificity. Influenza-specific DR*0401 CD4 T(CM) were isolated from the peripheral blood of normal donors by flow cytometry. Peptide-loaded aAPC were not sufficient to induce resting CD4 T(CM) to proliferate. In contrast, we found that the beads efficiently promoted the growth of previously activated CD4 T(CM) cells, yielding cultures with >80% Ag-specific CD4 cells after two stimulations. Further stimulation with peptide-loaded aAPC increased purity to >99% Ag-specific T cells. After in vitro culture for 3-12 wk, the flu-specific CD4 T(CM) had surface markers that were generally consistent with an effector phenotype described for CD8 T cells, except for the maintenance of CD28 expression. The T(CM) were capable of 20-40 mean population doublings in vitro, and the expanded cells produced IFN-gamma, IL-2, and TNF-alpha in response to Ag, and a subset of cells also secreted IL-4 with PMA/ionomycin treatment. In conclusion, aAPCs expand T(CM) that have extensive replicative capacity, and have potential applications in adoptive immunotherapy as well as for studying the biology of human MHC class II-restricted T cells.
Collapse
Affiliation(s)
- Marcela V Maus
- Abramson Family Cancer Research Institute, University of Pennsylvania Cancer Center,University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Taner SB, Onfelt B, Pirinen NJ, McCann FE, Magee AI, Davis DM. Control of Immune Responses by Trafficking Cell Surface Proteins, Vesicles and Lipid Rafts to and from the Immunological Synapse. Traffic 2004; 5:651-61. [PMID: 15296490 DOI: 10.1111/j.1600-0854.2004.00214.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Supramolecular clusters at the immunological synapse provide a mechanism for structuring complex communication networks between cells of the immune system. Regulating intra- and intercellular trafficking of proteins and lipids to and from the immunological synapse provides an additional level of complexity in determining the functional outcome of immune cell interactions. An emergent principle is that molecules requiring tightly regulated cell surface expression, e.g. negative regulators of cell activation or molecules promoting cytotoxicity, are trafficked to the immunological synapse from intracellular secretory as required lysosomes. Many molecules required for the early stages of the intercellular communication are already present at the cell surface, sometimes in lipid rafts, and are rapidly translocated laterally to the intercellular contact. Our understanding of these events critically depends on utilizing appropriate technologies for probing supramolecular recognition in live cells. Thus, we also present here a critical discussion of the technologies used to study lipid rafts and, more broadly, a map of the spatial and temporal dimensions covered by current live cell physical techniques, highlighting where advances are needed to exceed current spatial and temporal boundaries.
Collapse
Affiliation(s)
- Sabrina B Taner
- Department of Biological Sciences, Imperial College London, SW7 2AZ, UK
| | | | | | | | | | | |
Collapse
|
36
|
O'Keefe JP, Blaine K, Alegre ML, Gajewski TF. Formation of a central supramolecular activation cluster is not required for activation of naive CD8+ T cells. Proc Natl Acad Sci U S A 2004; 101:9351-6. [PMID: 15192143 PMCID: PMC438980 DOI: 10.1073/pnas.0305965101] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Although both naive and effector T lymphocytes interact with antigen-expressing cells, the functional outcome of these interactions is distinct. Naive CD8(+) T cells are activated to proliferate and differentiate into effector cytolytic T lymphocytes (CTL), whereas CTL interact with specific targets, such as tumor cells, to induce apoptotic death. We recently observed that several molecules linked to actin cytoskeleton dynamics were up-regulated in effector vs. naive CD8(+) T cells, leading us to investigate whether T cell differentiation is accompanied by changes in actin-dependent processes. We observed that both naive and effector CD8(+) T cells underwent T cell receptor capping and formed stable conjugates with antigen-specific antigen-presenting cells. However, the characteristics of the immunological synapse were distinct. Whereas accumulation of signaling molecules at the T cell/antigen-presenting cell contact site was detectable in both naive and effector CD8(+) T cells, only effector cells developed a central supramolecular activation cluster as defined by punctate focusing of PKC theta, phospho-PKC theta, and phospho-ZAP70. Extended kinetics, CD28 costimulation, and high-affinity antigenic peptide did not promote PKC theta focusing in naive cells. Nonetheless, naive CD8(+) T cells polarized the microtubule organizing center, produced IL-2, proliferated, and differentiated into effector cells. Our results suggest that the formation of a central supramolecular activation cluster is not required for activation of naive CD8(+) T cells and support the notion that one role of an organized immune synapse is directed delivery of effector function.
Collapse
Affiliation(s)
- James P O'Keefe
- Committee on Cancer Biology and Department of Pathology, University of Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
37
|
Djordjevic JT, Schibeci SD, Stewart GJ, Williamson P. HIV type 1 Nef increases the association of T cell receptor (TCR)-signaling molecules with T cell rafts and promotes activation-induced raft fusion. AIDS Res Hum Retroviruses 2004; 20:547-55. [PMID: 15186530 DOI: 10.1089/088922204323087804] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
HIV-1 Nef (Nef) is a myristoylated protein that contributes to HIV disease pathogenesis. Nef has a modulatory effect on T cell receptor (TCR) signaling, resulting in up-regulation of interleukin-2 (IL-2) production in stimulated T cells. Recent studies have shown that efficient TCR signaling requires enhanced association of TCR-signaling molecules with plasma membrane microdomains (lipid rafts) and fusion of rafts into larger structures. We utilized Jurkat T cell lines expressing wild-type Nef (Nef(wt)) and a myristoylation-deficient form of Nef (Nef(G)2(A)), from an inducible promoter, to determine the effects of Nef on the association of TCR-signaling molecules with rafts in nonstimulated T cells. In addition, the effect of Nef on raft size, before and after TCR activation by CD3 cross-linking, was also examined. Following induction, Nef(wt) was associated with both rafts and nonrafts, while Nef(G)2(A) was almost exclusively cytosolic. Induction of Nef(wt), but not Nef(G)2(A), coincided with an increased association of the src family tyrosine kinase, Lck, and TCRzeta with rafts, but not with nonrafts. Further, rafts were found to be significantly larger in CD3-activated T cells in the presence of Nef(wt) when compared to nonexpressing cells. We propose that myristoylated, raft-localized Nef primes resting T cells for activation by increasing the levels of signaling molecules within rafts, and that TCR activation is enhanced by the capacity of Nef to promote raft fusion.
Collapse
Affiliation(s)
- Julianne T Djordjevic
- Institute for Immunology and Allergy Research, Westmead Millennium Institute, NSW 2145, Australia
| | | | | | | |
Collapse
|
38
|
de Mello Coelho V, Nguyen D, Giri B, Bunbury A, Schaffer E, Taub DD. Quantitative differences in lipid raft components between murine CD4+ and CD8+ T cells. BMC Immunol 2004; 5:2. [PMID: 15005797 PMCID: PMC343273 DOI: 10.1186/1471-2172-5-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2003] [Accepted: 01/30/2004] [Indexed: 11/26/2022] Open
Abstract
Background Lipid rafts have been shown to play a role in T cell maturation, activation as well as in the formation of immunological synapses in CD4+ helper and CD8+ cytotoxic T cells. However, the differential expression of lipid raft components between CD4+ and CD8+ T cells is still poorly defined. To examine this question, we analyzed the expression of GM1 in T cells from young and aged mice as well as the expression of the glycosylphosphatidylinositol (GPI)-linked protein Thy-1 and cholesterol in murine CD4+ and CD8+ T cell subpopulations. Results We found that CD4+CD8- and CD8+CD4- thymocytes at different stages of maturation display distinct GM1 surface expression. This phenomenon did not change with progressive aging, as these findings were consistent over the lifespan of the mouse. In the periphery, CD8+ T cells express significantly higher levels of GM1 than CD4+ T cells. In addition, we observed that GM1 levels increase over aging on CD8+ T cells but not in CD4+ T cells. We also verified that naïve (CD44lo) and memory (CD44hi) CD8+ T cells as well as naïve and memory CD4+ T cells express similar levels of GM1 on their surface. Furthermore, we found that CD8+ T cells express higher levels of the GPI-anchored cell surface protein Thy-1 associated with lipid raft domains as compared to CD4+ T cells. Finally, we observed higher levels of total cellular cholesterol in CD8+ T cells than CD4+ T cells. Conclusion These results demonstrate heterogeneity of lipid raft components between CD4+ and CD8+ T cells in young and aged mice. Such differences in lipid raft composition may contribute to the differential CD4 and CD8 molecule signaling pathways as well as possibly to the effector responses mediated by these T cell subsets following TCR activation.
Collapse
Affiliation(s)
- Valeria de Mello Coelho
- Laboratory of Immunology, Gerontology Research Center, National Institute on Aging, NIH, Baltimore, MD, 21224-6825, USA
| | - Dzung Nguyen
- Laboratory of Immunology, Gerontology Research Center, National Institute on Aging, NIH, Baltimore, MD, 21224-6825, USA
| | - Banabihari Giri
- Laboratory of Immunology, Gerontology Research Center, National Institute on Aging, NIH, Baltimore, MD, 21224-6825, USA
| | - Allyson Bunbury
- Laboratory of Immunology, Gerontology Research Center, National Institute on Aging, NIH, Baltimore, MD, 21224-6825, USA
| | - Eric Schaffer
- Laboratory of Immunology, Gerontology Research Center, National Institute on Aging, NIH, Baltimore, MD, 21224-6825, USA
| | - Dennis D Taub
- Laboratory of Immunology, Gerontology Research Center, National Institute on Aging, NIH, Baltimore, MD, 21224-6825, USA
| |
Collapse
|
39
|
Starr TK, Daniels MA, Lucido MM, Jameson SC, Hogquist KA. Thymocyte sensitivity and supramolecular activation cluster formation are developmentally regulated: a partial role for sialylation. THE JOURNAL OF IMMUNOLOGY 2004; 171:4512-20. [PMID: 14568924 DOI: 10.4049/jimmunol.171.9.4512] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
TCR reactivity is tuned during thymic development. Immature thymocytes respond to low-affinity self-ligands resulting in positive selection. Following differentiation, T cells no longer respond to low-affinity ligands, but respond well to high-affinity (foreign) ligands. We show in this study that this response includes integrin activation, supramolecular activation cluster formation, Ca(2+) flux, and CD69 expression. Because glycosylation patterns are known to change during T cell development, we tested whether alterations in sialylation influence CD8 T cell sensitivity to low affinity TCR ligands. Using neuraminidase treatment or genetic deficiency in the ST3Gal-I sialyltransferase, we show that desialylation of mature CD8 T cells enhances their sensitivity to low-affinity ligands, although these treatments do not completely recapitulate the dynamic range of immature T cells. These studies identify sialylation as one of the factors that regulate CD8 T cell tuning during development.
Collapse
Affiliation(s)
- Timothy K Starr
- Center for Immunology, Laboratory of Medicine and Pathology, University of Minnesota, Minneapolis MN 55455, USA
| | | | | | | | | |
Collapse
|
40
|
Shibuya K, Shirakawa J, Kameyama T, Honda SI, Tahara-Hanaoka S, Miyamoto A, Onodera M, Sumida T, Nakauchi H, Miyoshi H, Shibuya A. CD226 (DNAM-1) is involved in lymphocyte function-associated antigen 1 costimulatory signal for naive T cell differentiation and proliferation. ACTA ACUST UNITED AC 2004; 198:1829-39. [PMID: 14676297 PMCID: PMC2194159 DOI: 10.1084/jem.20030958] [Citation(s) in RCA: 200] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Upon antigen recognition by the T cell receptor, lymphocyte function–associated antigen 1 (LFA-1) physically associates with the leukocyte adhesion molecule CD226 (DNAM-1) and the protein tyrosine kinase Fyn. We show that lentiviral vector-mediated mutant (Y-F322) CD226 transferred into naive CD4+ helper T cells (Ths) inhibited interleukin (IL)-12–independent Th1 development initiated by CD3 and LFA-1 ligations. Moreover, proliferation induced by LFA-1 costimulatory signal was suppressed in mutant (Y-F322) CD226-transduced naive CD4+ and CD8+ T cells in the absence of IL-2. These results suggest that CD226 is involved in LFA-1–mediated costimulatory signals for triggering naive T cell differentiation and proliferation. We also demonstrate that although LFA-1, CD226, and Fyn are polarized at the immunological synapse upon stimulation with anti-CD3 in CD4+ and CD8+ T cells, lipid rafts are polarized in CD4+, but not CD8+, T cells. Moreover, proliferation initiated by LFA-1 costimulatory signal is suppressed by lipid raft disruption in CD4+, but not CD8+, T cells, suggesting that the LFA-1 costimulatory signal is independent of lipid rafts in CD8+ T cells.
Collapse
Affiliation(s)
- Kazuko Shibuya
- Laboratory for Immune Receptor, RIKEN Research Center for Allergy and Immunology, 3-1-1 Koyadai, Ibaraki 305-0074, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Araki H, Katayama N, Yamashita Y, Mano H, Fujieda A, Usui E, Mitani H, Ohishi K, Nishii K, Masuya M, Minami N, Nobori T, Shiku H. Reprogramming of human postmitotic neutrophils into macrophages by growth factors. Blood 2003; 103:2973-80. [PMID: 15070673 DOI: 10.1182/blood-2003-08-2742] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
It is generally recognized that postmitotic neutrophils give rise to polymorphonuclear neutrophils alone. We obtained evidence for a lineage switch of human postmitotic neutrophils into macrophages in culture. When the CD15+CD14- cell population, which predominantly consists of band neutrophils, was cultured with granulocyte macrophage-colony-stimulating factor, tumor necrosis factor-alpha, interferon-gamma, and interleukin-4, and subsequently with macrophage colony-stimulating factor alone, the resultant cells had morphologic, cytochemical, and phenotypic features of macrophages. In contrast to the starting population, they were negative for myeloperoxidase, specific esterase, and lactoferrin, and they up-regulated nonspecific esterase activity and the expression of macrophage colony-stimulating factor receptor, mannose receptor, and HLA-DR. CD15+CD14- cells proceeded to macrophages through the CD15-CD14- cell population. Microarray analysis of gene expression also disclosed the lineage conversion from neutrophils to macrophages. Macrophages derived from CD15+CD14- neutrophils had phagocytic function. Data obtained using 3 different techniques, including Ki-67 staining, bromodeoxyuridine incorporation, and cytoplasmic dye labeling, together with the yield of cells, indicated that the generation of macrophages from CD15+CD14- neutrophils did not result from a contamination of progenitors for macrophages. Our data show that in response to cytokines, postmitotic neutrophils can become macrophages. This may represent another differentiation pathway toward macrophages in human postnatal hematopoiesis.
Collapse
Affiliation(s)
- Hiroto Araki
- Second Department of Internal Medicine, Mie University School of Medicine, Tsu, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kupzig S, Korolchuk V, Rollason R, Sugden A, Wilde A, Banting G. Bst-2/HM1.24 is a raft-associated apical membrane protein with an unusual topology. Traffic 2003; 4:694-709. [PMID: 12956872 DOI: 10.1034/j.1600-0854.2003.00129.x] [Citation(s) in RCA: 354] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
An expression screen of a rat cDNA library for sequences encoding Golgi-localized integral membrane proteins identified a protein with an apparent novel topology, i.e. with both an N-terminal transmembrane domain and a C-terminal glycosyl-phosphatidylinositol (GPI) anchor. Our data are consistent with this. Thus, the protein would have a topology that, in mammalian cells, is shared only by a minor, but pathologically important, topological isoform of the prion protein (PrP). The human orthologue of this protein has been described previously (BST-2 or HM1.24 antigen) as a cell surface molecule that appears to be involved in early pre-B-cell development and which is present at elevated levels at the surface of myeloma cells. We show that rat BST-2/HM1.24 has both a cell surface and an intracellular (juxtanuclear) location and is efficiently internalized from the cell surface. We also show that the cell surface pool of BST-2/HM1.24 is predominantly present in the apical plasma membrane of polarized cells. The fact that rat BST-2/HM1.24 apparently possesses a GPI anchor led us to speculate that it might exist in cholesterol-rich lipid microdomains (lipid rafts) at the plasma membrane. Data from several experiments are consistent with this localization. We present a model in which BST-2/HM1.24 serves to link adjacent lipid rafts within the plasma membrane.
Collapse
Affiliation(s)
- Sabine Kupzig
- Department of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | | | | | | | | |
Collapse
|
43
|
Hare KJ, Pongracz J, Jenkinson EJ, Anderson G. Modeling TCR signaling complex formation in positive selection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:2825-31. [PMID: 12960303 DOI: 10.4049/jimmunol.171.6.2825] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
T cell receptor signaling in the thymus can result in positive selection, and hence progressive maturation to the CD4(+)8(-) or CD4(-)8(+) stage, or induction of apoptosis by negative selection. Although it is poorly understood how TCR ligation at the CD4(+)8(+) stage can lead to such different cell fates, it is thought that the strength of signal may play a role in determining the outcome of TCR signaling. In this study, we have characterized the formation of an active signaling complex in thymocytes undergoing positive selection as a result of interaction with thymic epithelial cells. Although this signaling complex involves redistribution of cell surface and intracellular molecules, reminiscent of that observed in T cell activation, accumulation of GM1-containing lipid rafts was not observed. However, enforced expression of the costimulatory molecule CD80 on thymic epithelium induced GM1 polarization in thymocytes, and was accompanied by reduced positive selection and increased apoptosis. We suggest that the presence or absence of CD80 costimulation influences the outcome of TCR signaling in CD4(+)8(+) thymocytes through differential lipid raft recruitment, thus determining overall signal strength and influencing developmental cell fate.
Collapse
MESH Headings
- Animals
- Antigens, CD
- CD3 Complex/metabolism
- Cell Aggregation/genetics
- Cell Aggregation/immunology
- Cell Communication/genetics
- Cell Communication/immunology
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Membrane/genetics
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Cell Separation
- Epithelial Cells/cytology
- Epithelial Cells/immunology
- G(M1) Ganglioside/metabolism
- Intracellular Fluid/immunology
- Intracellular Fluid/metabolism
- Leukocyte Common Antigens/metabolism
- Leukosialin
- Major Histocompatibility Complex/physiology
- Membrane Microdomains/genetics
- Membrane Microdomains/immunology
- Membrane Microdomains/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Models, Immunological
- Organ Culture Techniques
- Receptors, Antigen, T-Cell, alpha-beta/deficiency
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/physiology
- Sialoglycoproteins/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
Collapse
Affiliation(s)
- Katherine J Hare
- Department of Anatomy, Medical Research Council Centre for Immune Regulation, University of Birmingham, Birmingham, United Kingdom.
| | | | | | | |
Collapse
|
44
|
Sanui T, Inayoshi A, Noda M, Iwata E, Oike M, Sasazuki T, Fukui Y. DOCK2 is essential for antigen-induced translocation of TCR and lipid rafts, but not PKC-theta and LFA-1, in T cells. Immunity 2003; 19:119-29. [PMID: 12871644 DOI: 10.1016/s1074-7613(03)00169-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
DOCK2 is a mammalian homolog of Caenorhabditis elegans CED-5 and Drosophila melanogaster Myoblast City which are known to regulate actin cytoskeleton. DOCK2 is critical for lymphocyte migration, yet the role of DOCK2 in TCR signaling remains unclear. We show here that DOCK2 is essential for TCR-mediated Rac activation and immunological synapse formation. In DOCK2-deficient T cells, antigen-induced translocation of TCR and lipid rafts, but not PKC-theta and LFA-1, to the APC interface was severely impaired, resulting in a significant reduction of antigen-specific T cell proliferation. In addition, we found that the efficacy of both positive and negative selection was reduced in DOCK2-deficient mice. These results suggest that DOCK2 regulates T cell responsiveness through remodeling of actin cytoskeleton via Rac activation.
Collapse
Affiliation(s)
- Terukazu Sanui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | | | | | | | | | | | | |
Collapse
|