1
|
Moore EK, Strazza M, Hu X, Tymm C, Paiola M, Shannon MJ, Xie X, Bukhari S, Lerrer S, Mace EM, Winchester R, Mor A. PAG orchestrates T cell immune synapse function by binding to actin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.29.625440. [PMID: 39677716 PMCID: PMC11642767 DOI: 10.1101/2024.11.29.625440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Many immunotherapies impact T cell function by impacting the immune synapse. While immunotherapy is extremely successful in some patients, in many others, it fails to help or causes complications, including immune-related adverse events. Phosphoprotein Associated with Glycosphingolipid Rich Microdomains 1 (PAG) is a transmembrane scaffold protein with importance in T cell signaling. PAG has 10 tyrosine phosphorylation sites where many kinases and phosphatases bind. PAG is palmitoylated, so it localizes in lipid rafts of the membrane, and contains a C-terminal PDZ domain to link to the actin cytoskeleton. As a link between signaling-protein-rich membrane regions and the actin cytoskeleton, PAG is an exciting and novel target for manipulating immune function. Here, we sought to determine if PAG works with actin to control T cell synapse organization and function. We found that PAG and actin dynamics are tightly coordinated during synapse maturation. A PDZ domain mutation disrupts the PAG-actin interaction, significantly impairing synapse formation, stability, and function. To assess the impact of the PDZ mutation functionally in vivo, we employed a mouse model of type IV hypersensitivity and an OVA-tumor mouse model. In both systems, mice with T cells expressing PDZ-mutant PAG had diminished immune responses, including impaired cytotoxic function. These findings highlight the importance of the PAG-actin link for effective T cell immune synapse formation and function. The results of our study suggest that targeting PAG is a promising approach for modulating immune responses and treating immune-related diseases. One Sentence Summary Adaptor protein PAG links to the actin cytoskeleton, and this link is essential for T cell synapse formation and cytotoxic function.
Collapse
|
2
|
Shao D, Bai T, Zhu B, Guo X, Dong K, Shi J, Huang Q, Kong J. Construction and Mechanism of IL-15-Based Coactivated Polymeric Micelles for NK Cell Immunotherapy. Adv Healthc Mater 2024; 13:e2302589. [PMID: 37897328 DOI: 10.1002/adhm.202302589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/12/2023] [Indexed: 10/30/2023]
Abstract
Natural killer (NK) cells are an important contributor to cancer immunotherapy, but their antitumor efficacy remains suboptimal. While cytokine-based priming shows promise in enhancing NK-cell activity, its clinical translation faces many challenges, including coactivation of multiple cytokines, poor pharmacokinetics, and limited mechanistic understanding. Here, this work develops a polymeric micelle-based IL-15/IL-2 codelivery system (IL-15/2-PEG-PTMC) for NK-cell activation. In vivo studies demonstrate that half-life of IL-15 and IL-2 and the recruitment of NK cell within tumor tissue are significantly increased after PEG-PTMC loading. Coupled with the coactivation effect of IL-15 and IL-2 conferred by this system, it noticeably delays the growth of tumors compared to conventional NK-cell activation approach, that is free IL-15 and IL-2. It is also surprisingly found that cholesterol metabolism is highly involved in the NK cell activation by IL-15/2-PEG-PTMC. Following stimulation with IL-15/2-PEG-PTMC or IL-15, NK cells undergo a series of cholesterol metabolism reprogramming, which elevates the cholesterol levels on NK cell membrane. This in turn promotes the formation of lipid rafts and activates immune synapses, effectively contributing to the enhancement of NK cell's antitumor activity. It is believed that it will open a new avenue for improving the efficacy of NK cell immunotherapy by regulating cholesterol metabolism.
Collapse
Affiliation(s)
- Dongyan Shao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Ting Bai
- Key Laboratory of Textile Fiber and Products, Ministry of Education, Wuhan Textile University, Wuhan, 430200, China
| | - Bobo Zhu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Xiaojia Guo
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Kai Dong
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Junling Shi
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Qingsheng Huang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jie Kong
- Shaanxi Key Laboratory of Macromolecular Science and Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| |
Collapse
|
3
|
Vallés AS, Barrantes FJ. The synaptic lipidome in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:184033. [PMID: 35964712 DOI: 10.1016/j.bbamem.2022.184033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 06/15/2023]
Abstract
Adequate homeostasis of lipid, protein and carbohydrate metabolism is essential for cells to perform highly specific tasks in our organism, and the brain, with its uniquely high energetic requirements, posesses singular characteristics. Some of these are related to its extraordinary dotation of synapses, the specialized subcelluar structures where signal transmission between neurons occurs in the central nervous system. The post-synaptic compartment of excitatory synapses, the dendritic spine, harbors key molecules involved in neurotransmission tightly packed within a minute volume of a few femtoliters. The spine is further compartmentalized into nanodomains that facilitate the execution of temporo-spatially separate functions in the synapse. Lipids play important roles in this structural and functional compartmentalization and in mechanisms that impact on synaptic transmission. This review analyzes the structural and dynamic processes involving lipids at the synapse, highlighting the importance of their homeostatic balance for the physiology of this complex and highly specialized structure, and underscoring the pathologies associated with disbalances of lipid metabolism, particularly in the perinatal and late adulthood periods of life. Although small variations of the lipid profile in the brain take place throughout the adult lifespan, the pathophysiological consequences are clinically manifested mostly during late adulthood. Disturbances in lipid homeostasis in the perinatal period leads to alterations during nervous system development, while in late adulthood they favor the occurrence of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ana Sofia Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (UNS-CONICET), 8000 Bahía Blanca, Argentina.
| | - Francisco J Barrantes
- Laboratory of Molecular Neurobiology, Institute of Biomedical Research (BIOMED), UCA-CONICET, Av. Alicia Moreau de Justo 1600, Buenos Aires C1107AAZ, Argentina.
| |
Collapse
|
4
|
Moore EK, Strazza M, Mor A. Combination Approaches to Target PD-1 Signaling in Cancer. Front Immunol 2022; 13:927265. [PMID: 35911672 PMCID: PMC9330480 DOI: 10.3389/fimmu.2022.927265] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer remains the second leading cause of death in the US, accounting for 25% of all deaths nationwide. Immunotherapy techniques bolster the immune cells' ability to target malignant cancer cells and have brought immense improvements in the field of cancer treatments. One important inhibitory protein in T cells, programmed cell death protein 1 (PD-1), has become an invaluable target for cancer immunotherapy. While anti-PD-1 antibody therapy is extremely successful in some patients, in others it fails or even causes further complications, including cancer hyper-progression and immune-related adverse events. Along with countless translational studies of the PD-1 signaling pathway, there are currently close to 5,000 clinical trials for antibodies against PD-1 and its ligand, PD-L1, around 80% of which investigate combinations with other therapies. Nevertheless, more work is needed to better understand the PD-1 signaling pathway and to facilitate new and improved evidence-based combination strategies. In this work, we consolidate recent discoveries of PD-1 signaling mediators and their therapeutic potential in combination with anti-PD-1/PD-L1 agents. We focus on the phosphatases SHP2 and PTPN2; the kinases ITK, VRK2, GSK-3, and CDK4/6; and the signaling adaptor protein PAG. We discuss their biology both in cancer cells and T cells, with a focus on their role in relation to PD-1 to determine their potential in therapeutic combinations. The literature discussed here was obtained from a search of the published literature and ClinicalTrials.gov with the following key terms: checkpoint inhibition, cancer immunotherapy, PD-1, PD-L1, SHP2, PTPN2, ITK, VRK2, CDK4/6, GSK-3, and PAG. Together, we find that all of these proteins are logical and promising targets for combination therapy, and that with a deeper mechanistic understanding they have potential to improve the response rate and decrease adverse events when thoughtfully used in combination with checkpoint inhibitors.
Collapse
Affiliation(s)
- Emily K. Moore
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
| | - Marianne Strazza
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
| | - Adam Mor
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
5
|
Yan K, Meng Q, He H, Zhu H, Wang Z, Han L, Huang Q, Zhang Z, Yawalkar N, Zhou H, Xu J. iTRAQ-based quantitative proteomics reveals biomarkers/pathways in psoriasis that can predict the efficacy of methotrexate. J Eur Acad Dermatol Venereol 2022; 36:1784-1795. [PMID: 35666151 DOI: 10.1111/jdv.18292] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/05/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Methotrexate (MTX) is the first-line medicine to treat psoriasis. So far, there has been less research on protein biomarkers to predict its efficacy by the proteomic technique. OBJECTIVES To evaluate differentially expressed proteins in peripheral mononuclear cells (PBMCs) between good responders (GRs) and non-responders (NRs) after MTX treatment, compared with normal controls (NCs). METHODS We quantified protein expression of PBMCs with 4 GRs and 4 NRs to MTX and 4 NCs by isobaric tags for relative and absolute quantification (iTRAQ), analyzing and identifying proteins related to efficacy of MTX in 18 psoriatic patients. RESULTS A total of 3,177 proteins had quantitative information, and 403 differentially expressed proteins (fold change ≥ 1.2, p < .05) were identified. Compared to NCs, upregulated proteins (ANXA6, RPS27A, EZR, XRCC6), participating in the activation of NF-κB, the JAK-STAT pathway, and neutrophil degranulation were detected in GRs. The proteins (GPV, FN1, STOM), involving platelet activation, signaling and aggregation as well as neutrophil degranulation were significantly downregulated in GRs. These proteins returned to normal levels after MTX treatment. Furthermore, Western blotting identified the expression of ANXA6 and STAT1 in PBMCs, which were significantly downregulated in GRs, but not in NRs. CONCLUSIONS We identified seven differentially expressed and regulated proteins (ANXA6, GPV, FN1, XRCC6, STOM, RPS27A, and EZR) as biomarkers to predict MTX efficacy in NF-κB signaling, JAK-STAT pathways, neutrophil degranulation, platelet activation, signaling and aggregation.
Collapse
Affiliation(s)
- Kexiang Yan
- Institute of Dermatology and Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qian Meng
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Han He
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hongwen Zhu
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhicheng Wang
- Department of Clinical Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Ling Han
- Institute of Dermatology and Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qiong Huang
- Institute of Dermatology and Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zhenghua Zhang
- Institute of Dermatology and Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Nikhil Yawalkar
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Hu Zhou
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jinhua Xu
- Institute of Dermatology and Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| |
Collapse
|
6
|
Han J, Zhang H, Li N, Aziz AUR, Zhang Z, Liu B. The raft cytoskeleton binding protein complexes personate functional regulators in cell behaviors. Acta Histochem 2022; 124:151859. [PMID: 35123353 DOI: 10.1016/j.acthis.2022.151859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/23/2022] [Accepted: 01/27/2022] [Indexed: 12/08/2022]
Abstract
Several cytoskeleton proteins interact with raft proteins to form raft-cytoskeleton binding protein complexes (RCPCs) that control cell migration and adhesion. The purpose of this paper is to review the latest research on the modes and mechanisms by which a RCPC controls different cellular functions. This paper discusses RCPC composition and its role in cytoskeleton reorganization, as well as the latest developments in molecular mechanisms that regulate cell adhesion and migration under normal conditions. In addition, the role of some external stimuli (such as stress and chemical signals) in this process is further debated, and meanwhile potential mechanisms for RCPC to regulate lipid raft fluidity is proposed. Thus, this review mainly contributes to the understanding of RCPC signal transduction in cells. Additionally, the targeted signal transduction of RCPC and its mechanism connection with cell behaviors will provide a logical basis for the development of unified interventions to combat metastasis related dysfunction and diseases.
Collapse
Affiliation(s)
- Jinxin Han
- School of Biomedical Engineering, Dalian University of Technology, Key Laboratory for Integrated Circuit and Biomedical Electronic System of Liaoning Province, Dalian 116024, China
| | - Hangyu Zhang
- School of Biomedical Engineering, Dalian University of Technology, Key Laboratory for Integrated Circuit and Biomedical Electronic System of Liaoning Province, Dalian 116024, China
| | - Na Li
- School of Biomedical Engineering, Dalian University of Technology, Key Laboratory for Integrated Circuit and Biomedical Electronic System of Liaoning Province, Dalian 116024, China
| | - Aziz Ur Rehman Aziz
- School of Biomedical Engineering, Dalian University of Technology, Key Laboratory for Integrated Circuit and Biomedical Electronic System of Liaoning Province, Dalian 116024, China
| | - Zhengyao Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| | - Bo Liu
- School of Biomedical Engineering, Dalian University of Technology, Key Laboratory for Integrated Circuit and Biomedical Electronic System of Liaoning Province, Dalian 116024, China.
| |
Collapse
|
7
|
Vallés AS, Barrantes FJ. Dendritic spine membrane proteome and its alterations in autistic spectrum disorder. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:435-474. [PMID: 35034726 DOI: 10.1016/bs.apcsb.2021.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Dendritic spines are small protrusions stemming from the dendritic shaft that constitute the primary specialization for receiving and processing excitatory neurotransmission in brain synapses. The disruption of dendritic spine function in several neurological and neuropsychiatric diseases leads to severe information-processing deficits with impairments in neuronal connectivity and plasticity. Spine dysregulation is usually accompanied by morphological alterations to spine shape, size and/or number that may occur at early pathophysiological stages and not necessarily be reflected in clinical manifestations. Autism spectrum disorder (ASD) is one such group of diseases involving changes in neuronal connectivity and abnormal morphology of dendritic spines on postsynaptic neurons. These alterations at the subcellular level correlate with molecular changes in the spine proteome, with alterations in the copy number, topography, or in severe cases in the phenotype of the molecular components, predominantly of those proteins involved in spine recognition and adhesion, reflected in abnormally short lifetimes of the synapse and compensatory increases in synaptic connections. Since cholinergic neurotransmission participates in the regulation of cognitive function (attention, memory, learning processes, cognitive flexibility, social interactions) brain acetylcholine receptors are likely to play an important role in the dysfunctional synapses in ASD, either directly or indirectly via the modulatory functions exerted on other neurotransmitter receptor proteins and spine-resident proteins.
Collapse
Affiliation(s)
- Ana Sofía Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (UNS-CONICET), Bahía Blanca, Argentina
| | - Francisco J Barrantes
- Instituto de Investigaciones Biomédicas (BIOMED), UCA-CONICET, Buenos Aires, Argentina.
| |
Collapse
|
8
|
Strazza M, Azoulay-Alfaguter I, Peled M, Adam K, Mor A. Transmembrane adaptor protein PAG is a mediator of PD-1 inhibitory signaling in human T cells. Commun Biol 2021; 4:672. [PMID: 34083754 PMCID: PMC8175585 DOI: 10.1038/s42003-021-02225-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
The inhibitory receptor PD-1 is expressed on T cells to inhibit select functions when ligated. The complete signaling mechanism downstream of PD-1 has yet to be uncovered. Here, we discovered phosphoprotein associated with glycosphingolipid-enriched microdomains 1 (PAG) is phosphorylated following PD-1 ligation and associate this with inhibitory T cell function. Clinical cohort analysis correlates low PAG expression with increased survival from numerous tumor types. PAG knockdown in T cells prevents PD-1-mediated inhibition of cytokine secretion, cell adhesion, CD69 expression, and ERK204/187 phosphorylation, and enhances phosphorylation of SRC527 following PD-1 ligation. PAG overexpression rescues these effects. In vivo, PAG contributes greatly to the growth of two murine tumors, MC38 and B16, and limits T cell presence within the tumor. Moreover, PAG deletion sensitizes tumors to PD-1 blockade. Here PAG is established as a critical mediator of PD-1 signaling and as a potential target to enhance T cell activation in tumors.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Antigens, CD/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Cell Line, Tumor
- Cells, Cultured
- Cytokines/metabolism
- Humans
- Lectins, C-Type/metabolism
- Lymphocyte Activation
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Phosphorylation
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/metabolism
- Signal Transduction
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Mice
Collapse
Affiliation(s)
- Marianne Strazza
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | | | - Michael Peled
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Kieran Adam
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Adam Mor
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA.
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
9
|
Mastrogiovanni M, Juzans M, Alcover A, Di Bartolo V. Coordinating Cytoskeleton and Molecular Traffic in T Cell Migration, Activation, and Effector Functions. Front Cell Dev Biol 2020; 8:591348. [PMID: 33195256 PMCID: PMC7609836 DOI: 10.3389/fcell.2020.591348] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/24/2020] [Indexed: 12/28/2022] Open
Abstract
Dynamic localization of receptors and signaling molecules at the plasma membrane and within intracellular vesicular compartments is crucial for T lymphocyte sensing environmental cues, triggering membrane receptors, recruiting signaling molecules, and fine-tuning of intracellular signals. The orchestrated action of actin and microtubule cytoskeleton and intracellular vesicle traffic plays a key role in all these events that together ensure important steps in T cell physiology. These include extravasation and migration through lymphoid and peripheral tissues, T cell interactions with antigen-presenting cells, T cell receptor (TCR) triggering by cognate antigen-major histocompatibility complex (MHC) complexes, immunological synapse formation, cell activation, and effector functions. Cytoskeletal and vesicle traffic dynamics and their interplay are coordinated by a variety of regulatory molecules. Among them, polarity regulators and membrane-cytoskeleton linkers are master controllers of this interplay. Here, we review the various ways the T cell plasma membrane, receptors, and their signaling machinery interplay with the actin and microtubule cytoskeleton and with intracellular vesicular compartments. We highlight the importance of this fine-tuned crosstalk in three key stages of T cell biology involving cell polarization: T cell migration in response to chemokines, immunological synapse formation in response to antigen cues, and effector functions. Finally, we discuss two examples of perturbation of this interplay in pathological settings, such as HIV-1 infection and mutation of the polarity regulator and tumor suppressor adenomatous polyposis coli (Apc) that leads to familial polyposis and colorectal cancer.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
- Collège Doctoral, Sorbonne Université, Paris, France
| | - Marie Juzans
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Andrés Alcover
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Vincenzo Di Bartolo
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| |
Collapse
|
10
|
Ullah MA, Vicente CT, Collinson N, Curren B, Sikder MAA, Sebina I, Simpson J, Varelias A, Lindquist JA, Ferreira MAR, Phipps S. PAG1 limits allergen-induced type 2 inflammation in the murine lung. Allergy 2020; 75:336-345. [PMID: 31321783 DOI: 10.1111/all.13991] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/30/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Phosphoprotein associated with glycosphingolipid-enriched microdomains 1 (PAG1) is a transmembrane adaptor protein that affects immune receptor signaling in T and B cells. Evidence from genome-wide association studies of asthma suggests that genetic variants that regulate the expression of PAG1 are associated with asthma risk. However, it is not known whether PAG1 expression is causally related to asthma pathophysiology. Here, we investigated the role of PAG1 in a preclinical mouse model of house dust mite (HDM)-induced allergic sensitization and allergic airway inflammation. METHODS Pag1-deficient (Pag1-/- ) and wild-type (WT) mice were sensitized or sensitized/challenged to HDM, and hallmark features of allergic inflammation were assessed. The contribution of T cells was assessed through depletion (anti-CD4 antibody) and adoptive transfer studies. RESULTS Type 2 inflammation (eosinophilia, eotaxin-2 expression, IL-4/IL-5/IL-13 production, mucus production) in the airways and lungs was significantly increased in HDM sensitized/challenged Pag1-/- mice compared to WT mice. The predisposition to allergic sensitization was associated with increased airway epithelial high-mobility group box 1 (HMGB1) translocation and release, increased type 2 innate lymphoid cells (ILC2s) and monocyte-derived dendritic cell numbers in the mediastinal lymph nodes, and increased T-helper type 2 (TH 2)-cell differentiation. CD4+ T-cell depletion studies or the adoptive transfer of WT OVA-specific CD4+ T cells to WT or Pag1-/- recipients demonstrated that the heightened propensity for TH 2-cell differentiation was both T cell intrinsic and extrinsic. CONCLUSION PAG1 deficiency increased airway epithelial activation, ILC2 expansion, and TH 2 differentiation. As a consequence, PAG1 deficiency predisposed toward allergic sensitization and increased the severity of experimental asthma.
Collapse
Affiliation(s)
- Md Ashik Ullah
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
| | - Cristina T. Vicente
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
| | | | - Bodie Curren
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
| | - Md Al Amin Sikder
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
| | - Ismail Sebina
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
| | - Jennifer Simpson
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
| | - Jonathan A. Lindquist
- Clinic for Nephrology and Hypertension, Diabetology and Endocrinology Otto‐von‐Guericke University Magdeburg Germany
| | | | - Simon Phipps
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
- Australian Infectious Diseases Research Centre University of Queensland Brisbane Qld Australia
| |
Collapse
|
11
|
Pera T, Tompkins E, Katz M, Wang B, Deshpande DA, Weinman EJ, Penn RB. Specificity of NHERF1 regulation of GPCR signaling and function in human airway smooth muscle. FASEB J 2019; 33:9008-9016. [PMID: 31042404 PMCID: PMC6662985 DOI: 10.1096/fj.201900323r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/08/2019] [Indexed: 12/21/2022]
Abstract
Na+/H+ exchanger regulatory factor 1 (NHERF1; also known as ezrin-radixin-moesin-binding phosphoprotein 50) is a PSD-95, disc large, zona occludens-1 adapter that acts as a scaffold for signaling complexes and cytoskeletal-plasma membrane interactions. NHERF1 is crucial to β-2-adrenoceptor (β2AR)-mediated activation of cystic fibrosis transmembrane conductance regulator (CFTR) in epithelial cells, and NHERF1 has been proposed to mediate the recycling of internalized β2AR back to the cell membrane. In the current study, we assessed the role of NHERF1 in regulating cAMP-mediated signaling and immunomodulatory functions in airway smooth muscle (ASM). NHERF1 knockdown attenuated the induction of (protein kinase A) phospho-vasodilator-stimulated phosphoprotein (p-VASP) by isoproterenol (ISO), prostaglandin E2 (PGE2), or forskolin (FSK) as well as the induction of p-heat shock protein 20 after 4 h of stimulation with ISO and FSK. NHERF1 knockdown fully abrogated the ISO-, PGE2-, and FSK-induced IL-6 gene expression and cytokine production without affecting cAMP-mediated phosphodiesterase 4D (PDE4D) gene expression, phospho-cAMP response element-binding protein (p-CREB), and cAMP response element (CRE)-Luc, or PDGF-induced cyclin D1 expression. Interestingly, NHERF1 knockdown prevented ISO-induced chromatin-binding of the transcription factor CCAAT-enhancer-binding protein-β (c/EBPβ). c/EBPβ knockdown almost completely abrogated the cAMP-mediated IL-6 but not PDE4D gene expression. The differential regulation of cAMP-induced signaling and gene expression in our study indicates a role for NHERF1 in the compartmentalization of cAMP signaling in ASM.-Pera, T., Tompkins, E., Katz, M., Wang, B., Deshpande, D. A., Weinman, E. J., Penn, R. B. Specificity of NHERF1 regulation of GPCR signaling and function in human airway smooth muscle.
Collapse
Affiliation(s)
- Tonio Pera
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, The Jane and Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Eric Tompkins
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, The Jane and Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Michael Katz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, The Jane and Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Bin Wang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, The Jane and Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Deepak A. Deshpande
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, The Jane and Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Edward J. Weinman
- Department of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Raymond B. Penn
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, The Jane and Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Sun S, Adyshev D, Dudek S, Paul A, McColloch A, Cho M. Cholesterol-dependent Modulation of Stem Cell Biomechanics: Application to Adipogenesis. J Biomech Eng 2019; 141:2729412. [PMID: 30901381 DOI: 10.1115/1.4043253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Indexed: 11/08/2022]
Abstract
Cell mechanics has been shown to regulate stem cell differentiation. We have previously reported that altered cell stiffness of mesenchymal stem cells can delay or facilitate biochemically directed differentiation. One of the factors that can affect the cell stiffness is cholesterol. However, the effect of cholesterol on differentiation of human mesenchymal stem cells (hMSCs) remains elusive. In this paper, we demonstrate that cholesterol is involved in the modulation of the cell stiffness and subsequent adipogenic differentiation. Rapid cytoskeletal actin reorganization was evident and correlated with the cell's Young's modulus measured using atomic force microscopy (AFM). In addition, the level of membrane-bound cholesterol was found to increase during adipogenic differentiation and inversely varied with the cell stiffness. Furthermore, cholesterol played a key role in the regulation of the cell morphology and biomechanics, suggesting its crucial involvement in mechanotransduction. To better understand the underlying mechanisms, we investigated the effect of cholesterol on the membrane-cytoskeleton linker proteins (ezrin and moesin). Cholesterol depletion was found to up-regulate the ezrin expression which promoted cell spreading, increased Young's modulus, and hindered adipogenesis. In contrast, cholesterol enrichment increased the moesin expression, decreased Young's modulus, and induced cell rounding and facilitated adipogenesis. Taken together, cholesterol appears to regulate the stem cell mechanics and adipogenesis through the membrane-associated linker proteins.
Collapse
Affiliation(s)
- Shan Sun
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607
| | - Djanybek Adyshev
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607
| | - Steve Dudek
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607
| | - Amit Paul
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607
| | - Andrew McColloch
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019
| | - Michael Cho
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019
| |
Collapse
|
13
|
Cong BB, Gao MH, Li B, Wang B, Zhang B, Wang LN, Zhang SC, Li HQ, Wang Z, Han SY. Overexpression of Csk-binding protein/phosphoprotein associated with glycosphingolipid-enriched microdomains induces cluster of differentiation 59-mediated apoptosis in Jurkat cells. Exp Ther Med 2018; 15:4139-4148. [PMID: 29725363 PMCID: PMC5920370 DOI: 10.3892/etm.2018.5940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 10/25/2017] [Indexed: 11/17/2022] Open
Abstract
Csk-binding protein/phosphoprotein associated with glycosphingolipid-enriched microdomains (CBP/PAG) is a membrane-bound adaptor protein that downregulates the activation of Src family kinases present in lipid rafts. To elucidate the role of CBP/PAG in human T cell activation, a cell line overexpressing CBP/PAG was constructed and the function of CBP/PAG in Jurkat cells was examined. The present study revealed that increased CBP/PAG expression in T cells significantly enhanced their apoptosis and reduced cellular activation and proliferation. Overexpression of CBP/PAG suppressed the growth of Jurkat cells by recruiting c-Src and its negative regulator, C-terminal Src kinase (CSK), to lipid rafts. The negative regulation of CBP/PAG was enhanced in the presence of anti-cluster of differentiation (CD)59 monoclonal antibodies. In addition, a significant association was revealed between the location of CBP/PAG and CD59, which were co-expressed in the same region of the cell membrane, implicating a potential overlap of the elicited signaling pathways. These results indicate that CBP/PAG functions as a negative regulator of cell signal transduction and suggest that CD59 may strengthen the role of negative feedback regulation.
Collapse
Affiliation(s)
- Bei-Bei Cong
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Mei-Hua Gao
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Bing Li
- Department of Genetics, Medical College of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Bing Wang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Bei Zhang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Li-Na Wang
- Department of Blood Transfusion, Shandong Qilu Hospital, Qingdao, Shandong 266071, P.R. China
| | - Shu-Chao Zhang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Hua-Qiao Li
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Zhong Wang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Shu-Yi Han
- Medical Research and Laboratory Diagnostic Center, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250000, P.R. China
| |
Collapse
|
14
|
Arumugham VB, Ulivieri C, Onnis A, Finetti F, Tonello F, Ladant D, Baldari CT. Compartmentalized Cyclic AMP Production by the Bordetella pertussis and Bacillus anthracis Adenylate Cyclase Toxins Differentially Affects the Immune Synapse in T Lymphocytes. Front Immunol 2018; 9:919. [PMID: 29765373 PMCID: PMC5938339 DOI: 10.3389/fimmu.2018.00919] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/13/2018] [Indexed: 01/01/2023] Open
Abstract
A central feature of the immune synapse (IS) is the tight compartmentalization of membrane receptors and signaling mediators that is functional for its ability to coordinate T cell activation. Second messengers centrally implicated in this process, such as Ca2+ and diacyl glycerol, also undergo compartmentalization at the IS. Current evidence suggests a more complex scenario for cyclic AMP (cAMP), which acts both as positive and as negative regulator of T-cell antigen receptor (TCR) signaling and which, as such, must be subjected to a tight spatiotemporal control to allow for signaling at the IS. Here, we have used two bacterial adenylate cyclase toxins that produce cAMP at different subcellular localizations as the result of their distinct routes of cell invasion, namely Bordetella pertussis CyaA and Bacillus anthracis edema toxin (ET), to address the ability of the T cell to confine cAMP to the site of production and to address the impact of compartmentalized cAMP production on IS assembly and function. We show that CyaA, which produces cAMP close to the synaptic membrane, affects IS stability by modulating not only the distribution of LFA-1 and its partners talin and L-plastin, as previously partly reported but also by promoting the sustained synaptic accumulation of the A-kinase adaptor protein ezrin and protein kinase A while suppressing the β-arrestin-mediated recruitment of phosphodiesterase 4B. These effects are dependent on the catalytic activity of the toxin and can be reproduced by treatment with a non-hydrolyzable cAMP analog. Remarkably, none of these effects are elicited by ET, which produces cAMP at a perinuclear localization, despite its ability to suppress TCR signaling and T cell activation through its cAMP-elevating activity. These results show that the IS responds solely to local elevations of cAMP and provide evidence that potent compartmentalization mechanisms are operational in T cells to contain cAMP close to the site of production, even when produced at supraphysiological levels.
Collapse
Affiliation(s)
| | | | - Anna Onnis
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | - Fiorella Tonello
- Neuroscience Institute, Italian National Research Council, Padua, Italy
| | - Daniel Ladant
- Department of Structural Biology and Chemistry, Institut Pasteur, Paris, France
| | | |
Collapse
|
15
|
Miszczuk GS, Barosso IR, Larocca MC, Marrone J, Marinelli RA, Boaglio AC, Sánchez Pozzi EJ, Roma MG, Crocenzi FA. Mechanisms of canalicular transporter endocytosis in the cholestatic rat liver. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1072-1085. [DOI: 10.1016/j.bbadis.2018.01.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/12/2018] [Accepted: 01/16/2018] [Indexed: 01/03/2023]
|
16
|
Agarwal S, Ghosh R, Chen Z, Lakoma A, Gunaratne PH, Kim ES, Shohet JM. Transmembrane adaptor protein PAG1 is a novel tumor suppressor in neuroblastoma. Oncotarget 2018; 7:24018-26. [PMID: 26993602 PMCID: PMC5029681 DOI: 10.18632/oncotarget.8116] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 03/01/2016] [Indexed: 02/06/2023] Open
Abstract
(NB) is the most common extracranial pediatric solid tumor with high mortality rates. The tyrosine kinase c-Src has been known to play an important role in differentiation of NB cells, but the mechanism of c-Src regulation has not been defined. Here, we characterize PAG1 (Cbp, Csk binding protein), a central inhibitor of c-Src and other Src family kinases, as a novel tumor suppressor in NB. Clinical cohort analysis demonstrate that low expression of PAG1 is a significant prognostic factor for high stage disease, increased relapse, and worse overall survival for children with NB. PAG1 knockdown in NB cells promotes proliferation and anchorage-independent colony formation with increased activation of AKT and ERK downstream of c-Src, while PAG1 overexpression significantly rescues these effects. In vivo, PAG1 overexpression significantly inhibits NB tumorigenicity in an orthotopic xenograft model. Our results establish PAG1 as a potent tumor suppressor in NB by inhibiting c-Src and downstream effector pathways. Thus, reactivation of PAG1 and inhibition of c-Src kinase activity represents an important novel therapeutic approach for high-risk NB.
Collapse
Affiliation(s)
- Saurabh Agarwal
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, and Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Rajib Ghosh
- Department of Biology & Biochemistry, University of Houston, Houston, Texas 77204, USA
| | - Zaowen Chen
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, and Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Anna Lakoma
- Michael E. DeBakey, Department of Surgery, Division of Pediatric Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Preethi H Gunaratne
- Department of Biology & Biochemistry, University of Houston, Houston, Texas 77204, USA
| | - Eugene S Kim
- Michael E. DeBakey, Department of Surgery, Division of Pediatric Surgery, Baylor College of Medicine, Houston, Texas 77030, USA.,Department of Surgery, Division of Pediatric Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California 90027, USA
| | - Jason M Shohet
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer Center, and Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
17
|
Tóth EA, Oszvald Á, Péter M, Balogh G, Osteikoetxea-Molnár A, Bozó T, Szabó-Meleg E, Nyitrai M, Derényi I, Kellermayer M, Yamaji T, Hanada K, Vígh L, Matkó J. Nanotubes connecting B lymphocytes: High impact of differentiation-dependent lipid composition on their growth and mechanics. Biochim Biophys Acta Mol Cell Biol Lipids 2017. [PMID: 28645851 DOI: 10.1016/j.bbalip.2017.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nanotubes (NTs) are thin, long membranous structures forming novel, yet poorly known communication pathways between various cell types. Key mechanisms controlling their growth still remained poorly understood. Since NT-forming capacity of immature and mature B cells was found largely different, we investigated how lipid composition and molecular order of the membrane affect NT-formation. Screening B cell lines with various differentiation stages revealed that NT-growth linearly correlates with membrane ganglioside levels, while it shows maximum as a function of cholesterol level. NT-growth of B lymphocytes is promoted by raftophilic phosphatidylcholine and sphingomyelin species, various glycosphingolipids, and docosahexaenoic acid-containing inner leaflet lipids, through supporting membrane curvature, as demonstrated by comparative lipidomic analysis of mature versus immature B cell membranes. Targeted modification of membrane cholesterol and sphingolipid levels altered NT-forming capacity confirming these findings, and also highlighted that the actual lipid raft number may control NT-growth via defining the number of membrane-F-actin coupling sites. Atomic force microscopic mechano-manipulation experiments further proved that mechanical properties (elasticity or bending stiffness) of B cell NTs also depend on the actual membrane lipid composition. Data presented here highlight importance of the lipid side in controlling intercellular, nanotubular, regulatory communications in the immune system.
Collapse
Affiliation(s)
- Eszter A Tóth
- Department of Immunology, Eötvös Lorand University, Budapest, Hungary
| | - Ádám Oszvald
- Department of Immunology, Eötvös Lorand University, Budapest, Hungary
| | - Mária Péter
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Gábor Balogh
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary.
| | | | - Tamás Bozó
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Edina Szabó-Meleg
- Department of Biophysics, Medical Faculty, University of Pécs, Pécs, Hungary; Department of Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Miklós Nyitrai
- Department of Biophysics, Medical Faculty, University of Pécs, Pécs, Hungary; Department of Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Imre Derényi
- Department of Biological Physics, Eötvös Lorand University, Budapest, Hungary
| | - Miklós Kellermayer
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary; MTA-SE Molecular Biophysics Research Group, Semmelweis University, Budapest, Hungary
| | - Toshiyuki Yamaji
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shunjuku-ku, Tokyo, Japan
| | - Kentaro Hanada
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shunjuku-ku, Tokyo, Japan
| | - László Vígh
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | - János Matkó
- Department of Immunology, Eötvös Lorand University, Budapest, Hungary.
| |
Collapse
|
18
|
Dunst J, Azzouz N, Liu X, Tsukita S, Seeberger PH, Kamena F. Interaction between Plasmodium Glycosylphosphatidylinositol and the Host Protein Moesin Has No Implication in Malaria Pathology. Front Cell Infect Microbiol 2017; 7:183. [PMID: 28560184 PMCID: PMC5432536 DOI: 10.3389/fcimb.2017.00183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/27/2017] [Indexed: 11/17/2022] Open
Abstract
Glycosylphosphatidylinositol (GPI) anchor of Plasmodium falciparum origin is considered an important toxin leading to severe malaria pathology through stimulation of pro-inflammatory responses from innate immune cells. Even though the GPI-induced immune response is widely described to be mediated by pattern recognition receptors such as TLR2 and TLR4, previous studies have revealed that these two receptors are dispensable for the development of severe malaria pathology. Therefore, this study aimed at the identification of potential alternative Plasmodium GPI receptors. Herein, we have identified the host protein moesin as an interaction partner of Plasmodium GPI in vitro. Given previous reports indicating the relevance of moesin especially in the LPS-mediated induction of pro-inflammatory responses, we have conducted a series of in vitro and in vivo experiments to address the physiological relevance of the moesin-Plasmodium GPI interaction in the context of malaria pathology. We report here that although moesin and Plasmodium GPI interact in vitro, moesin is not critically involved in processes leading to Plasmodium-induced pro-inflammatory immune responses or malaria-associated cerebral pathology.
Collapse
Affiliation(s)
- Josefine Dunst
- Institute of Chemistry and Biochemistry, Free University BerlinBerlin, Germany
- Parasitology Unit, Max Planck Institute for Infection BiologyBerlin, Germany
| | - Nahid Azzouz
- Institute of Chemistry and Biochemistry, Free University BerlinBerlin, Germany
- Department of Biomolecular Systems, Max Planck Institute for Colloids and InterfacesPotsdam, Germany
| | - Xinyu Liu
- Department of Chemistry, University of PittsburghPittsburgh, PA, USA
| | - Sachiko Tsukita
- Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka UniversityOsaka, Japan
| | - Peter H. Seeberger
- Institute of Chemistry and Biochemistry, Free University BerlinBerlin, Germany
- Department of Biomolecular Systems, Max Planck Institute for Colloids and InterfacesPotsdam, Germany
| | - Faustin Kamena
- Institute of Chemistry and Biochemistry, Free University BerlinBerlin, Germany
- Parasitology Unit, Max Planck Institute for Infection BiologyBerlin, Germany
| |
Collapse
|
19
|
Astrocyte-to-neuron communication through integrin-engaged Thy-1/CBP/Csk/Src complex triggers neurite retraction via the RhoA/ROCK pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:243-254. [DOI: 10.1016/j.bbamcr.2016.11.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/27/2016] [Accepted: 11/07/2016] [Indexed: 12/22/2022]
|
20
|
Vaquero J, Nguyen Ho-Bouldoires TH, Clapéron A, Fouassier L. Role of the PDZ-scaffold protein NHERF1/EBP50 in cancer biology: from signaling regulation to clinical relevance. Oncogene 2017; 36:3067-3079. [PMID: 28068322 DOI: 10.1038/onc.2016.462] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/26/2016] [Accepted: 10/31/2016] [Indexed: 12/14/2022]
Abstract
The transmission of cellular information requires fine and subtle regulation of proteins that need to interact in a coordinated and specific way to form efficient signaling networks. The spatial and temporal coordination relies on scaffold proteins. Thanks to protein interaction domains such as PDZ domains, scaffold proteins organize multiprotein complexes enabling the proper transmission of cellular information through intracellular networks. NHERF1/EBP50 is a PDZ-scaffold protein that was initially identified as an organizer and regulator of transporters and channels at the apical side of epithelia through actin-binding ezrin-moesin-radixin proteins. Since, NHERF1/EBP50 has emerged as a major regulator of cancer signaling network by assembling cancer-related proteins. The PDZ-scaffold EBP50 carries either anti-tumor or pro-tumor functions, two antinomic functions dictated by EBP50 expression or subcellular localization. The dual function of NHERF1/EBP50 encompasses the regulation of several major signaling pathways engaged in cancer, including the receptor tyrosine kinases PDGFR and EGFR, PI3K/PTEN/AKT and Wnt-β-catenin pathways.
Collapse
Affiliation(s)
- J Vaquero
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,FONDATION ARC, Villejuif, France
| | - T H Nguyen Ho-Bouldoires
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,FONDATION ARC, Villejuif, France
| | - A Clapéron
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - L Fouassier
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| |
Collapse
|
21
|
Wehbi VL, Taskén K. Molecular Mechanisms for cAMP-Mediated Immunoregulation in T cells - Role of Anchored Protein Kinase A Signaling Units. Front Immunol 2016; 7:222. [PMID: 27375620 PMCID: PMC4896925 DOI: 10.3389/fimmu.2016.00222] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/23/2016] [Indexed: 12/20/2022] Open
Abstract
The cyclic AMP/protein kinase A (cAMP/PKA) pathway is one of the most common and versatile signal pathways in eukaryotic cells. A-kinase anchoring proteins (AKAPs) target PKA to specific substrates and distinct subcellular compartments providing spatial and temporal specificity for mediation of biological effects channeled through the cAMP/PKA pathway. In the immune system, cAMP is a potent negative regulator of T cell receptor-mediated activation of effector T cells (Teff) acting through a proximal PKA/Csk/Lck pathway anchored via a scaffold consisting of the AKAP Ezrin holding PKA, the linker protein EBP50, and the anchoring protein phosphoprotein associated with glycosphingolipid-enriched microdomains holding Csk. As PKA activates Csk and Csk inhibits Lck, this pathway in response to cAMP shuts down proximal T cell activation. This immunomodulating pathway in Teff mediates clinically important responses to regulatory T cell (Treg) suppression and inflammatory mediators, such as prostaglandins (PGs), adrenergic stimuli, adenosine, and a number of other ligands. A major inducer of T cell cAMP levels is PG E2 (PGE2) acting through EP2 and EP4 prostanoid receptors. PGE2 plays a crucial role in the normal physiological control of immune homeostasis as well as in inflammation and cancer immune evasion. Peripherally induced Tregs express cyclooxygenase-2, secrete PGE2, and elicit the immunosuppressive cAMP pathway in Teff as one tumor immune evasion mechanism. Moreover, a cAMP increase can also be induced by indirect mechanisms, such as intercellular transfer between T cells. Indeed, Treg, known to have elevated levels of intracellular cAMP, may mediate their suppressive function by transferring cAMP to Teff through gap junctions, which we speculate could also be regulated by PKA/AKAP complexes. In this review, we present an updated overview on the influence of cAMP-mediated immunoregulatory mechanisms acting through localized cAMP signaling and the therapeutical increasing prospects of AKAPs disruptors in T-cell immune function.
Collapse
Affiliation(s)
- Vanessa L. Wehbi
- Nordic EMBL Partnership, Centre for Molecular Medicine Norway, Oslo University Hospital, University of Oslo, Oslo, Norway
- Jebsen Inflammation Research Centre, Oslo University Hospital, Oslo, Norway
- Biotechnology Centre, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Kjetil Taskén
- Nordic EMBL Partnership, Centre for Molecular Medicine Norway, Oslo University Hospital, University of Oslo, Oslo, Norway
- Jebsen Inflammation Research Centre, Oslo University Hospital, Oslo, Norway
- Biotechnology Centre, Oslo University Hospital, University of Oslo, Oslo, Norway
- Jebsen Centre for Cancer Immunotherapy, Oslo University Hospital, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
22
|
Pore D, Gupta N. The ezrin-radixin-moesin family of proteins in the regulation of B-cell immune response. Crit Rev Immunol 2016; 35:15-31. [PMID: 25746045 DOI: 10.1615/critrevimmunol.2015012327] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Dynamic reorganization of the cortical cytoskeleton is essential for numerous cellular processes, including B- and T-cell activation and migration. The ezrin-radixin-moesin (ERM) family of proteins plays structural and regulatory roles in the rearrangement of plasma membrane flexibility and protrusions through its members' reversible interaction with cortical actin filaments and the plasma membrane. Recent studies demonstrated that ERM proteins not only are involved in cytoskeletal organization but also offer a platform for the transmission of signals in response to a variety of extracellular stimuli through their ability to cross-link transmembrane receptors with downstream signaling components. In this review, we summarize present knowledge relating to ERMs and recent progress made toward elucidating a novel role for them in the regulation of B-cell function in health and disease.
Collapse
Affiliation(s)
- Debasis Pore
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Neetu Gupta
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
23
|
Vicente C, Edwards S, Hillman K, Kaufmann S, Mitchell H, Bain L, Glubb D, Lee J, French J, Ferreira M. Long-Range Modulation of PAG1 Expression by 8q21 Allergy Risk Variants. Am J Hum Genet 2015. [PMID: 26211970 DOI: 10.1016/j.ajhg.2015.06.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The gene(s) whose expression is regulated by allergy risk variants is unknown for many loci identified through genome-wide association studies. Addressing this knowledge gap might point to new therapeutic targets for allergic disease. The aim of this study was to identify the target gene(s) and the functional variant(s) underlying the association between rs7009110 on chromosome 8q21 and allergies. Eight genes are located within 1 Mb of rs7009110. Multivariate association analysis of publicly available exon expression levels from lymphoblastoid cell lines (LCLs) identified a significant association between rs7009110 and the expression of a single gene, PAG1 (p = 0.0017), 732 kb away. Analysis of histone modifications and DNase I hypersensitive sites in LCLs identified four putative regulatory elements (PREs) in the region. Chromosome conformation capture confirmed that two PREs interacted with the PAG1 promoter, one in allele-specific fashion. To determine whether these PREs were functional, LCLs were transfected with PAG1 promoter-driven luciferase reporter constructs. PRE3 acted as a transcriptional enhancer for PAG1 exclusively when it carried the rs2370615:C allergy predisposing allele, a variant in complete linkage disequilibrium with rs7009110. As such, rs2370615, which overlaps RelA transcription factor (TF) binding in LCLs and was found to disrupt Foxo3a binding to PRE3, represents the putative functional variant in this locus. Our studies suggest that the risk-associated allele of rs2370615 predisposes to allergic disease by increasing PAG1 expression, which might promote B cell activation and have a pro-inflammatory effect. Inhibition of PAG1 expression or function might have therapeutic potential for allergic diseases.
Collapse
|
24
|
Moesin and stress-induced phosphoprotein-1 are possible sero-diagnostic markers of psoriasis. PLoS One 2014; 9:e101773. [PMID: 25010044 PMCID: PMC4092060 DOI: 10.1371/journal.pone.0101773] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 06/11/2014] [Indexed: 02/08/2023] Open
Abstract
To identify diagnostic markers for psoriasis vulgaris and psoriatic arthritis, autoantibodies in sera from psoriasis vulgaris and psoriatic arthritis patients were screened by two-dimensional immunoblotting (2D-IB). Based on 2D-IB and MADLI TOF/TOF-MS analyses, eleven proteins each in psoriasis vulgaris and psoriatic arthritis were identified as autoantigens. Furthermore, serum levels of moesin, keratin 17 (K17), annexin A1 (ANXA1), and stress-induced phophoprotein-1 (STIP1), which were detected as autoantigens, were studied by dot blot analysis with psoriasis patients and healthy controls. The levels of moesin and STIP1 were significantly higher in sera from patients with psoriasis vulgaris than in the controls (moesin: P<0.05, STIP1: P<0.005). The area under the curve (AUC) for moesin and STIP1 between patients with psoraisis vulgaris and controls was 0.747 and 0.792, respectively. STIP1 and K17 levels were significantly higher in sera from patients with psoriatic arthritis than in those with psoriasis vulgaris (P<0.05 each). The AUC for STIP1 and K17 between patients with psoriatic arthritis and psoriasis vulgaris was 0.69 and 0.72, respectively. The STIP1 or moesin, CK17 serum level was not correlated with disease activity of psoriasis patients. These data suggest that STIP1 and moesin may be novel and differential sero-diagnostic markers for psoriasis vulgaris and psoriatic arthritis.
Collapse
|
25
|
Palmitoylated transmembrane adaptor proteins in leukocyte signaling. Cell Signal 2014; 26:895-902. [PMID: 24440308 DOI: 10.1016/j.cellsig.2014.01.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/07/2014] [Accepted: 01/09/2014] [Indexed: 12/14/2022]
Abstract
Transmembrane adaptor proteins (TRAPs) are structurally related proteins that have no enzymatic function, but enable inducible recruitment of effector molecules to the plasma membrane, usually in a phosphorylation dependent manner. Numerous surface receptors employ TRAPs for either propagation or negative regulation of the signal transduction. Several TRAPs (LAT, NTAL, PAG, LIME, PRR7, SCIMP, LST1/A, and putatively GAPT) are known to be palmitoylated that could facilitate their localization in lipid rafts or tetraspanin enriched microdomains. This review summarizes expression patterns, binding partners, signaling pathways, and biological functions of particular palmitoylated TRAPs with an emphasis on the three most recently discovered members, PRR7, SCIMP, and LST1/A. Moreover, we discuss in silico methodology used for discovery of new family members, nature of their binding partners, and microdomain localization.
Collapse
|
26
|
|
27
|
Kvalvaag AS, Pust S, Sandvig K. Vps11, a subunit of the tethering complexes HOPS and CORVET, is involved in regulation of glycolipid degradation and retrograde toxin transport. Commun Integr Biol 2014; 7:e28129. [PMID: 24778763 PMCID: PMC3995726 DOI: 10.4161/cib.28129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/04/2014] [Accepted: 02/06/2014] [Indexed: 11/19/2022] Open
Abstract
We recently reported that ERM (ezrin, radixin, moesin) proteins are involved in intracellular sorting of Shiga toxin (Stx) and its receptor globotriaosylceramide (Gb3), and that depletion of ezrin and moesin reduced retrograde Golgi transport of Stx. In the same study, we found that knockdown of Vps11, a core subunit of both the homotypic fusion and protein sorting (HOPS) complex and the class C core vacuole/endosome tethering factor (CORVET), increased retrograde transport of Stx and could counteract the inhibiting effect of moesin and ezrin knockdown. In this study we demonstrate that Vps11 knockdown also leads to increased Stx toxicity as well as increased retrograde transport and toxicity of ricin. Additionally, we show that knockdown of Vps11 restores the reduced Gb3 level observed after moesin depletion.
Collapse
Affiliation(s)
- Audun Sverre Kvalvaag
- Department of Biochemistry; Institute for Cancer Research; The Norwegian Radium Hospital; Oslo University Hospital; Montebello, Oslo, Norway ; Department of Biosciences; Faculty of Mathematics and Natural Sciences; University of Oslo; Oslo, Norway ; Centre for Cancer Biomedicine; Faculty of Medicine; University of Oslo; Montebello, Oslo, Norway
| | - Sascha Pust
- Department of Biochemistry; Institute for Cancer Research; The Norwegian Radium Hospital; Oslo University Hospital; Montebello, Oslo, Norway ; Centre for Cancer Biomedicine; Faculty of Medicine; University of Oslo; Montebello, Oslo, Norway
| | - Kirsten Sandvig
- Department of Biochemistry; Institute for Cancer Research; The Norwegian Radium Hospital; Oslo University Hospital; Montebello, Oslo, Norway ; Department of Biosciences; Faculty of Mathematics and Natural Sciences; University of Oslo; Oslo, Norway ; Centre for Cancer Biomedicine; Faculty of Medicine; University of Oslo; Montebello, Oslo, Norway
| |
Collapse
|
28
|
Hrdinka M, Horejsi V. PAG - a multipurpose transmembrane adaptor protein. Oncogene 2013; 33:4881-92. [DOI: 10.1038/onc.2013.485] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 09/25/2013] [Accepted: 09/25/2013] [Indexed: 12/25/2022]
|
29
|
Parameswaran N, Gupta N. Re-defining ERM function in lymphocyte activation and migration. Immunol Rev 2013; 256:63-79. [DOI: 10.1111/imr.12104] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Neetha Parameswaran
- Department of Immunology; Lerner Research Institute; Cleveland Clinic; Cleveland OH USA
| | - Neetu Gupta
- Department of Immunology; Lerner Research Institute; Cleveland Clinic; Cleveland OH USA
| |
Collapse
|
30
|
Kvalvaag AS, Pust S, Sundet KI, Engedal N, Simm R, Sandvig K. The ERM proteins ezrin and moesin regulate retrograde Shiga toxin transport. Traffic 2013; 14:839-52. [PMID: 23593995 DOI: 10.1111/tra.12077] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 04/12/2013] [Accepted: 04/17/2013] [Indexed: 01/23/2023]
Abstract
The ERM proteins (ezrin, radixin and moesin) are known for connecting the actin cytoskeleton to the plasma membrane. They have been found to associate with lipid rafts as well as to be important for endosomal sorting and receptor signaling. However, little is known about the role of ERM proteins in retrograde transport and lipid homeostasis. In this study, we show that ezrin and moesin are important for efficient cell surface association of Shiga toxin (Stx) as well as for its retrograde transport. Furthermore, we show that depletion of these proteins influences endosomal dynamics and seems to enhance Stx transport toward lysosomes. We also show that knockdown of Vps11, a subunit of the HOPS complex, leads to increased retrograde Stx transport and reverses the inhibiting effect of ezrin and moesin knockdown. Importantly, retrograde transport of the plant toxin ricin, which binds to both glycolipids and glycoproteins with a terminal galactose, seems to be unaffected by ezrin and moesin depletion.
Collapse
Affiliation(s)
- Audun Sverre Kvalvaag
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, N-0379, Oslo, Norway
| | | | | | | | | | | |
Collapse
|
31
|
Yang J, Singh V, Cha B, Chen TE, Sarker R, Murtazina R, Jin S, Zachos NC, Patterson GH, Tse CM, Kovbasnjuk O, Li X, Donowitz M. NHERF2 protein mobility rate is determined by a unique C-terminal domain that is also necessary for its regulation of NHE3 protein in OK cells. J Biol Chem 2013; 288:16960-16974. [PMID: 23612977 DOI: 10.1074/jbc.m113.470799] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Na(+)/H(+) exchanger regulatory factor (NHERF) proteins are a family of PSD-95/Discs-large/ZO-1 (PDZ)-scaffolding proteins, three of which (NHERFs 1-3) are localized to the brush border in kidney and intestinal epithelial cells. All NHERF proteins are involved in anchoring membrane proteins that contain PDZ recognition motifs to form multiprotein signaling complexes. In contrast to their predicted immobility, NHERF1, NHERF2, and NHERF3 were all shown by fluorescence recovery after photobleaching/confocal microscopy to be surprisingly mobile in the microvilli of the renal proximal tubule OK cell line. Their diffusion coefficients, although different among the three, were all of the same magnitude as that of the transmembrane proteins, suggesting they are all anchored in the microvilli but to different extents. NHERF3 moves faster than NHERF1, and NHERF2 moves the slowest. Several chimeras and mutants of NHERF1 and NHERF2 were made to determine which part of NHERF2 confers the slower mobility rate. Surprisingly, the slower mobility rate of NHERF2 was determined by a unique C-terminal domain, which includes a nonconserved region along with the ezrin, radixin, moesin (ERM) binding domain. Also, this C-terminal domain of NHERF2 determined its greater detergent insolubility and was necessary for the formation of larger multiprotein NHERF2 complexes. In addition, this NHERF2 domain was functionally significant in NHE3 regulation, being necessary for stimulation by lysophosphatidic acid of activity and increased mobility of NHE3, as well as necessary for inhibition of NHE3 activity by calcium ionophore 4-Br-A23187. Thus, multiple functions of NHERF2 require involvement of an additional domain in this protein.
Collapse
Affiliation(s)
- Jianbo Yang
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Varsha Singh
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Boyoung Cha
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Tian-E Chen
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Rafiquel Sarker
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Rakhilya Murtazina
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Shi Jin
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Nicholas C Zachos
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - George H Patterson
- Biophotonics Section, National Institute of Biomedical Imaging and Bioengineering, NIH, Bethesda, Maryland 20892
| | - C Ming Tse
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Olga Kovbasnjuk
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Xuhang Li
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Mark Donowitz
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205; Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205.
| |
Collapse
|
32
|
Smida M, Cammann C, Gurbiel S, Kerstin N, Lingel H, Lindquist S, Simeoni L, Brunner-Weinzierl MC, Suchanek M, Schraven B, Lindquist JA. PAG/Cbp suppression reveals a contribution of CTLA-4 to setting the activation threshold in T cells. Cell Commun Signal 2013; 11:28. [PMID: 23601194 PMCID: PMC3763844 DOI: 10.1186/1478-811x-11-28] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 04/03/2013] [Indexed: 11/12/2022] Open
Abstract
Background PAG/Cbp represents a ubiquitous mechanism for regulating Src family kinases
by recruiting Csk to the plasma membrane, thereby controlling cellular
activation. Since Src kinases are known oncogenes, we used RNA interference
in primary human T cells to test whether the loss of PAG resulted in
lymphocyte transformation. Results PAG-depletion enhanced Src kinase activity and augmented proximal T-cell
receptor signaling; exactly the phenotype expected for loss of this negative
regulator. Surprisingly, rather than becoming hyper-proliferative,
PAG-suppressed T cells became unresponsive. This was mediated by a
Fyn-dependent hyper-phosphorylation of the inhibitory receptor CTLA-4, which
recruited the protein tyrosine phosphatase Shp-1 to lipid rafts.
Co-suppression of CTLA-4 abrogates this inhibition and restores
proliferation to T cells. Conclusion We have identified a fail-safe mechanism as well as a novel contribution of
CTLA-4 to setting the activation threshold in T cells.
Collapse
Affiliation(s)
- Michal Smida
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Leipziger Strasse 44, Magdeburg, 39120, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Voss JG, Dobra A, Morse C, Kovacs JA, Danner RL, Munson PJ, Logan C, Rangel Z, Adelsberger JW, McLaughlin M, Adams LD, Raju R, Dalakas MC. Fatigue-related gene networks identified in CD(14)+ cells isolated from HIV-infected patients: part I: research findings. Biol Res Nurs 2013; 15:137-51. [PMID: 23324479 DOI: 10.1177/1099800411421957] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE Human immunodeficiency virus (HIV)-related fatigue (HRF) is multicausal and potentially related to mitochondrial dysfunction caused by antiretroviral therapy with nucleoside reverse transcriptase inhibitors (NRTIs). METHODOLOGY The authors compared gene expression profiles of CD14(+) cells of low versus high fatigued, NRTI-treated HIV patients to healthy controls (n = 5/group). The authors identified 32 genes predictive of low versus high fatigue and 33 genes predictive of healthy versus HIV infection. The authors constructed genetic networks to further elucidate the possible biological pathways in which these genes are involved. RELEVANCE FOR NURSING PRACTICE: Genes including the actin cytoskeletal regulatory proteins Prokineticin 2 and Cofilin 2 along with mitochondrial inner membrane proteins are involved in multiple pathways and were predictors of fatigue status. Previously identified inflammatory and signaling genes were predictive of HIV status, clearly confirming our results and suggesting a possible further connection between mitochondrial function and HIV. Isolated CD14(+) cells are easily accessible cells that could be used for further study of the connection between fatigue and mitochondrial function of HIV patients. IMPLICATION FOR PRACTICE The findings from this pilot study take us one step closer to identifying biomarker targets for fatigue status and mitochondrial dysfunction. Specific biomarkers will be pertinent to the development of methodologies to diagnosis, monitor, and treat fatigue and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Joachim G Voss
- Biobehavioral Nursing & Health Systems Department, University of Washington, School of Nursing, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ke Q, Wu J, Ming B, Zhu S, Yu M, Wang Y, Hu C, Xu S, Luo Z. Identification of the PAG1 gene as a novel target of inherent radioresistance in human laryngeal carcinoma cells. Cancer Biother Radiopharm 2012; 27:678-84. [PMID: 22994656 DOI: 10.1089/cbr.2012.1191] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Laryngeal carcinoma, as a malignant tumor that occurs in the head and neck region, is widely treated by radiation, but in some cases, the cancer is radioresistant to the radiotherapy. The reason for the radioresistant response needs to be clinically understood. We designed our present study to identify the molecules that may be involved in this radioresistant response. In this study, we initially established the inherent radioresistant (Hep-2max) and radiosensitive (Hep-2min) cell lines from the parental laryngeal cancer cell line Hep-2. Furthermore, using microarray analysis, we identified a novel inherent radioresistance-related gene, phosphoprotein associated with glycosphingolipid-enriched microdomains1 (PAG1). We showed that siRNA directed against PAG1 in a radioresistant (Hep-2max) cell line dramatically enhanced the radiosensitivity and IR-induced cell death. On the contrary, ectopic expression of PAG1 in radiosensitive (Hep-2min) cell lines led to radioresistance and suppressed the IR-induced cell death. Taken together, our results indicate that the PAG1 gene may be a novel, promising radiosensitization target for laryngeal carcinoma.
Collapse
Affiliation(s)
- Qing Ke
- Department of Oncology, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan City, Hubei Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Yu W, Wang Y, Gong M, Pei F, Zheng J. Phosphoprotein associated with glycosphingolipid microdomains 1 inhibits the proliferation and invasion of human prostate cancer cells in vitro through suppression of Ras activation. Oncol Rep 2012; 28:606-14. [PMID: 22664862 DOI: 10.3892/or.2012.1848] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 03/26/2012] [Indexed: 11/06/2022] Open
Abstract
Phosphoprotein associated with glycosphingolipid microdomains 1 (PAG) is an important negative regulator of immune signaling in T lymphocytes. However, newly emerging evidence has indicated that PAG may play important roles in tumor cells. Our previously reported cDNA microarray experiments identified PAG as a gene down-regulated in the high metastatic potential prostate cancer cell line PC-3M-1E8. In this study, we investigated the role of PAG in the proliferation, invasion and metastasis of prostate cancer cells and the underlying mechanisms. We confirmed that the expression of PAG at both the mRNA and protein levels was low in PC-3M-1E8 and DU145 cells compared to low metastatic potential prostate cancer cells PC-3M-2B4. In addition, we demonstrated that the reintroduction of PAG to PC-3M-1E8 and DU145 cells led to reduced proliferation through cell cycle arrest, decreased anchorage-independent growth and reduced invasion ability of tumor cells in vitro. This is the first report demonstrating that PAG inhibits the proliferation and invasion potential of prostate cancer cells via the interaction with RasGAP to recruit RasGAP to the cell membrane, where RasGAP hydrolyzes GTP to GDP, reduces the level of activated Ras, and ultimately suppresses the activation of ERK1/2, cyclin D1 and other effectors of the Ras signaling pathway. Morphologically, we observed that PAG could diminish the formation of pseudopodia on the cell surface and redistribute the intracellular F-actin in PC-3M-1E8 cells, which directly leads to the decreased invasion and metastasis potential of tumor cells. Taken together, these results suggest that PAG acts to inhibit the development and metastasis of prostate cancers and represents a novel therapeutic target for prostate cancer.
Collapse
Affiliation(s)
- Wenjuan Yu
- Department of Pathology, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, PR China
| | | | | | | | | |
Collapse
|
36
|
Egea SC, Dickerson IM. Direct interactions between calcitonin-like receptor (CLR) and CGRP-receptor component protein (RCP) regulate CGRP receptor signaling. Endocrinology 2012; 153:1850-60. [PMID: 22315449 PMCID: PMC3320266 DOI: 10.1210/en.2011-1459] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Calcitonin gene-related peptide (CGRP) is a neuropeptide with multiple neuroendocrine roles, including vasodilation, migraine, and pain. The receptor for CGRP is a G protein-coupled receptor (GPCR) that requires three proteins for function. CGRP binds to a heterodimer composed of the GPCR calcitonin-like receptor (CLR) and receptor activity-modifying protein (RAMP1), a single transmembrane protein required for pharmacological specificity and trafficking of the CLR/RAMP1 complex to the cell surface. In addition, the CLR/RAMP1 complex requires a third protein named CGRP-receptor component protein (RCP) for signaling. Previous studies have demonstrated that depletion of RCP from cells inhibits CLR signaling, and in vivo studies have demonstrated that expression of RCP correlates with CLR signaling and CGRP efficacy. It is not known whether RCP interacts directly with CLR to exert its effect. The current studies identified a direct interaction between RCP and an intracellular domain of CLR using yeast two-hybrid analysis and coimmunoprecipitation. When this interacting domain of CLR was expressed as a soluble fusion protein, it coimmunoprecipitated with RCP and inhibited signaling from endogenous CLR. Expression of this dominant-negative domain of CLR did not significantly inhibit trafficking of CLR to the cell surface, and thus RCP may not have a chaperone function for CLR. Instead, RCP may regulate CLR signaling in the cell membrane, and direct interaction between RCP and CLR is required for CLR activation. To date, RCP has been found to interact only with CLR and represents a novel neuroendocrine regulatory step in GPCR signaling.
Collapse
Affiliation(s)
- Sophie C Egea
- Department of Physiology and Biophysics, University of Miami School of Medicine, Miami, Florida 33101, USA
| | | |
Collapse
|
37
|
Voss JG, Dobra A, Morse C, Kovacs JA, Raju R, Danner RL, Munson PJ, Logan C, Rangel Z, Adelsberger JW, McLaughlin M, Adams LD, Dalakas MC. Fatigue-related gene networks identified in CD14+ cells isolated from HIV-infected patients: part II: statistical analysis. Biol Res Nurs 2011; 15:152-9. [PMID: 22084402 DOI: 10.1177/1099800411423307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE In limited samples of valuable biological tissues, univariate ranking methods of microarray analyses often fail to show significant differences among expression profiles. In order to allow for hypothesis generation, novel statistical modeling systems can be greatly beneficial. The authors applied new statistical approaches to solve the issue of limited experimental data to generate new hypotheses in CD14(+) cells of patients with HIV-related fatigue (HRF) and healthy controls. METHODOLOGY We compared gene expression profiles of CD14(+) cells of nucleoside reverse transcriptase inhibitor (NRTI)-treated HIV patients with low versus high fatigue to healthy controls (n = 5 each). With novel Bayesian modeling procedures, the authors identified 32 genes predictive of low versus high fatigue and 33 genes predictive of healthy versus HIV infection. Sparse association and liquid association networks further elucidated the possible biological pathways in which these genes are involved. RELEVANCE FOR NURSING PRACTICE: Genetic networks developed in a comprehensive Bayesian framework from small sample sizes allow nursing researchers to design future research approaches to address such issues as HRF. IMPLICATION FOR PRACTICE The findings from this pilot study may take us one step closer to the development of useful biomarker targets for fatigue status. Specific and reliable tests are needed to diagnosis, monitor and treat fatigue and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Joachim G Voss
- Biobehavioral Nursing & Health Systems Department, School of Nursing, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kremer KN, Clift IC, Miamen AG, Bamidele AO, Qian NX, Humphreys TD, Hedin KE. Stromal cell-derived factor-1 signaling via the CXCR4-TCR heterodimer requires phospholipase C-β3 and phospholipase C-γ1 for distinct cellular responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:1440-7. [PMID: 21705626 PMCID: PMC3140596 DOI: 10.4049/jimmunol.1100820] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The CXCR4 chemokine receptor is a G protein-coupled receptor that signals in T lymphocytes by forming a heterodimer with the TCR. CXCR4 and TCR functions are consequently highly cross regulated, affecting T cell immune activation, cytokine secretion, and T cell migration. The CXCR4-TCR heterodimer stimulates T cell migration and activation of the ERK MAPK and downstream AP-1-dependent cytokine transcription in response to stromal cell-derived factor-1 (SDF-1), the sole chemokine ligand of CXCR4. These responses require Gi-type G proteins as well as TCR ITAM domains and the ZAP70 tyrosine kinase, thus indicating that the CXCR4-TCR heterodimer signals to integrate G protein-coupled receptor-associated and TCR-associated signaling molecules in response to SDF-1. Yet, the phospholipase C (PLC) isozymes responsible for coupling the CXCR4-TCR heterodimer to distinct downstream cellular responses are incompletely characterized. In this study, we demonstrate that PLC activity is required for SDF-1 to induce ERK activation, migration, and CXCR4 endocytosis in human T cells. SDF-1 signaling via the CXCR4-TCR heterodimer uses PLC-β3 to activate the Ras-ERK pathway and increase intracellular calcium ion concentrations, whereas PLC-γ1 is dispensable for these outcomes. In contrast, PLC-γ1, but not PLC-β3, is required for SDF-1-mediated migration via a mechanism independent of LAT. These results increase understanding of the signaling mechanisms employed by the CXCR4-TCR heterodimer, characterize new roles for PLC-β3 and PLC-γ1 in T cells, and suggest that multiple PLCs may also be activated downstream of other chemokine receptors to distinctly regulate migration versus other signaling functions.
Collapse
Affiliation(s)
- Kimberly N. Kremer
- Department of Immunology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Ian C. Clift
- Department of Immunology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Alexander G. Miamen
- Department of Immunology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Adebowale O. Bamidele
- Department of Immunology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN 55905
| | | | | | - Karen E. Hedin
- Department of Immunology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
39
|
Kim KB, Yi JS, Nguyen N, Lee JH, Kwon YC, Ahn BY, Cho H, Kim YK, Yoo HJ, Lee JS, Ko YG. Cell-surface receptor for complement component C1q (gC1qR) is a key regulator for lamellipodia formation and cancer metastasis. J Biol Chem 2011; 286:23093-101. [PMID: 21536672 DOI: 10.1074/jbc.m111.233304] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
We previously demonstrated that the receptor for the complement component C1q (gC1qR) is a lipid raft protein that is indispensable for adipogenesis and insulin signaling. Here, we provide the first report that gC1qR is an essential component of lamellipodia in human lung carcinoma A549 cells. Cell-surface gC1qR was concentrated in the lamellipodia along with CD44, monosialoganglioside, actin, and phosphorylated focal adhesion kinase in cells stimulated with insulin, IGF-1, EGF, or serum. The growth factor-induced lamellipodia formation and cell migration were significantly decreased in gC1qR-depleted cells, with a concomitant blunt activation of the focal adhesion kinase and the respective receptor tyrosine kinases. Moreover, the gC1qR-depleted cells exhibited a reduced proliferation rate in culture as well as diminished tumorigenic and metastatic activities in grafted mice. We therefore conclude that cell-surface gC1qR regulates lamellipodia formation and metastasis via receptor tyrosine kinase activation.
Collapse
Affiliation(s)
- Ki-Bum Kim
- College of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Francis SS, Sfakianos J, Lo B, Mellman I. A hierarchy of signals regulates entry of membrane proteins into the ciliary membrane domain in epithelial cells. ACTA ACUST UNITED AC 2011; 193:219-33. [PMID: 21444686 PMCID: PMC3082196 DOI: 10.1083/jcb.201009001] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The NHERF1-ERM-actin network comprises a selective retention matrix that prevents interacting membrane proteins from entering the ciliary membrane. The membrane of the primary cilium is continuous with the plasma membrane but compositionally distinct. Although some membrane proteins concentrate in the cilium, others such as podocalyxin/gp135 are excluded. We found that exclusion reflects a saturable selective retention mechanism. Podocalyxin is immobilized by its PDZ interaction motif binding to NHERF1 and thereby to the apical actin network via ERM family members. The retention signal was dominant, autonomous, and transferable to membrane proteins not normally excluded from the cilium. The NHERF1-binding domains of cystic fibrosis transmembrane conductance regulator and Csk-binding protein were also found to act as transferable retention signals. Addition of a retention signal could inhibit the ciliary localization of proteins (e.g., Smoothened) containing signals that normally facilitate concentration in the ciliary membrane. Proteins without a retention signal (e.g., green fluorescent protein–glycosylphosphatidylinositol) were found in the cilium, suggesting entry was not impeded by a diffusion barrier or lipid microdomain. Thus, a hierarchy of interactions controls the composition of the ciliary membrane, including selective retention, selective inclusion, and passive diffusion.
Collapse
Affiliation(s)
- Stephen S Francis
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | |
Collapse
|
41
|
Mosenden R, Singh P, Cornez I, Heglind M, Ruppelt A, Moutschen M, Enerbäck S, Rahmouni S, Taskén K. Mice with disrupted type I protein kinase A anchoring in T cells resist retrovirus-induced immunodeficiency. THE JOURNAL OF IMMUNOLOGY 2011; 186:5119-30. [PMID: 21430226 DOI: 10.4049/jimmunol.1100003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Type I protein kinase A (PKA) is targeted to the TCR-proximal signaling machinery by the A-kinase anchoring protein ezrin and negatively regulates T cell immune function through activation of the C-terminal Src kinase. RI anchoring disruptor (RIAD) is a high-affinity competitor peptide that specifically displaces type I PKA from A-kinase anchoring proteins. In this study, we disrupted type I PKA anchoring in peripheral T cells by expressing a soluble ezrin fragment with RIAD inserted in place of the endogenous A-kinase binding domain under the lck distal promoter in mice. Peripheral T cells from mice expressing the RIAD fusion protein (RIAD-transgenic mice) displayed augmented basal and TCR-activated signaling, enhanced T cell responsiveness assessed as IL-2 secretion, and reduced sensitivity to PGE(2)- and cAMP-mediated inhibition of T cell function. Hyperactivation of the cAMP-type I PKA pathway is involved in the T cell dysfunction of HIV infection, as well as murine AIDS, a disease model induced by infection of C57BL/6 mice with LP-BM5, a mixture of attenuated murine leukemia viruses. LP-BM5-infected RIAD-transgenic mice resist progression of murine AIDS and have improved viral control. This underscores the cAMP-type I PKA pathway in T cells as a putative target for therapeutic intervention in immunodeficiency diseases.
Collapse
Affiliation(s)
- Randi Mosenden
- The Biotechnology Center of Oslo, University of Oslo, N-0317 Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Garcia GG, Miller RA. Age-related defects in the cytoskeleton signaling pathways of CD4 T cells. Ageing Res Rev 2011; 10:26-34. [PMID: 19941976 DOI: 10.1016/j.arr.2009.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 11/18/2009] [Accepted: 11/18/2009] [Indexed: 11/25/2022]
Abstract
It has been postulated that the cytoskeleton controls many aspects of T cell function, including activation, proliferation and apoptosis. Recent advances in our understanding of F-actin polymerization and the Ezrin-Radixin-Moesin (ERM) family of cytoskeleton signal proteins have provided new insights into immunological synapse formation during T cell activation. During aging there is a significant decline of T cell function largely attributable to declines in activation of CD4 T cells and defects in the formation of the immunological synapse. Here we discuss recent progress in the understanding of how aging alters F-actin and ERM proteins in mouse CD4 T cells, and the implications of these changes for the T cell activation process.
Collapse
|
43
|
Cyclic AMP-mediated immune regulation--overview of mechanisms of action in T cells. Cell Signal 2010; 23:1009-16. [PMID: 21130867 DOI: 10.1016/j.cellsig.2010.11.018] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 11/23/2010] [Accepted: 11/25/2010] [Indexed: 01/21/2023]
Abstract
The canonical second messenger cAMP is well established as a potent negative regulator of T cell immune function. Through protein kinase A (PKA) it regulates T cell function at the level of transcription factors, members of the mitogen-activated protein kinase pathway, phospholipases (PLs), Ras homolog (Rho)A and proteins involved in the control of cell cycle progression. Type I PKA is the predominant PKA isoform in T cells. Furthermore, whereas type II PKA is located at the centrosome, type I PKA is anchored close to the T cell receptor (TCR) in lipid rafts by the Ezrin-ERM-binding phosphoprotein of 50 kDa (EBP50)-phosphoprotein associated with glycosphingolipid-enriched microdomains (PAG) scaffold complex. The most TCR-proximal target for type I PKA is C-terminal Src kinase (Csk), which upon activation by raft recruitment and phosphorylation inhibits the Src family tyrosine kinases Lck and Fyn and thus functions to maintain T cell homeostasis. Recently, induction of cAMP levels in responder T cells has emerged as one of the mechanisms by which regulatory T (T(R)) cells execute their suppressive action. Thus, the cAMP-type I PKA-Csk pathway emerges as a putative target for therapeutic intervention in autoimmune disorders as well as in cancer, where T(R) cell-mediated suppression contributes to suboptimal local immune responses.
Collapse
|
44
|
Lissner S, Nold L, Hsieh CJ, Turner JR, Gregor M, Graeve L, Lamprecht G. Activity and PI3-kinase dependent trafficking of the intestinal anion exchanger downregulated in adenoma depend on its PDZ interaction and on lipid rafts. Am J Physiol Gastrointest Liver Physiol 2010; 299:G907-20. [PMID: 20634435 PMCID: PMC5142450 DOI: 10.1152/ajpgi.00191.2010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The Cl/HCO(3) exchanger downregulated in adenoma (DRA) mediates electroneutral NaCl absorption in the intestine together with the apical Na/H exchanger NHE3. Lipid rafts (LR) modulate transport activity and are involved in phosphatidylinositol 3-kinase (PI3-kinase)-dependent trafficking of NHE3. Although DRA and NHE3 interact via PDZ adaptor proteins of the NHERF family, the role of LR and PDZ proteins in the regulation of DRA is unknown. We examined the association of DRA with LR using the nonionic detergent Triton X-100. DRA cofractionated with LR independently of its PDZ binding motif. Furthermore, DRA interacts with PDZK1, E3KARP, and IKEPP in LR, although their localization within lipid rafts is independent of DRA. Disruption of LR integrity resulted in the disappearance of DRA from LR, in a decrease of its surface expression and in a reduction of its activity. In HEK cells the inhibition of DRA by LR disruption was entirely dependent on the presence of the PDZ interaction motif. In addition, in Caco-2/BBE cells the inhibition by LR disruption was more pronounced in wild-type DRA than in mutated DRA (DRA-ETKFminus; lacking the PDZ binding motif)-expressing cells. Inhibition of PI3-kinase decreased the activity and the cell surface expression of wild-type DRA but not of DRA-ETKFminus; the partitioning into LR was unaffected. Furthermore, simultaneous inhibition of PI3-kinase and disruption of LR did not further decrease DRA activity and cell surface expression compared with LR disruption only. These results suggest that the activity of DRA depends on its LR association, on its PDZ interaction, and on PI3-kinase activity.
Collapse
Affiliation(s)
- S. Lissner
- 1st Medical Department, University of Tübingen, Tübingen, Germany,
| | - L. Nold
- 1st Medical Department, University of Tübingen, Tübingen, Germany,
| | - C.-J. Hsieh
- 1st Medical Department, University of Tübingen, Tübingen, Germany,
| | - J. R. Turner
- Department of Pathology, University of Chicago, Chicago, Illinois; and
| | - M. Gregor
- 1st Medical Department, University of Tübingen, Tübingen, Germany,
| | - L. Graeve
- Department of Biological Chemistry and Nutrition, University of Hohenheim, Hohenheim, Germany
| | - G. Lamprecht
- 1st Medical Department, University of Tübingen, Tübingen, Germany,
| |
Collapse
|
45
|
Transient and bilateral increase in Neuropilin-1, Fer kinase and collapsin response mediator proteins within membrane rafts following unilateral occlusion of the middle cerebral artery in mouse. Brain Res 2010; 1344:209-16. [PMID: 20493826 DOI: 10.1016/j.brainres.2010.05.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Revised: 05/10/2010] [Accepted: 05/12/2010] [Indexed: 01/22/2023]
Abstract
Membrane rafts, rich in sphingolipids and cholesterol, are membrane microdomains important in neuronal domain-specific signaling events such as during axonal outgrowth and neuronal death. The present study seeks to determine the spatiotemporal association of several axonal guidance signaling molecules with membrane rafts. These molecules are Neuropilin-1 (NRP-1), Fer Kinase, and collapsin response mediator proteins (CRMPs), which are known to have important functions in axonal outgrowth and neuronal death caused by cerebral ischemia. Mice were subjected to sham or a 1h unilateral middle cerebral artery occlusion (MCAO) followed by a time course of reperfusion up to 24h. Brain cortices were separated and membrane rafts were extracted based on their insolubility in Triton X-100 and separation by sucrose gradient fractionation. We demonstrate the early and transient induction of NRP-1 and CRMP-2 in membrane rafts in both ipsilateral and contralateral hemispheres, in contrast to an early, but sustained elevation of Fer kinase and other CRMPs (1, 3, 4, 5) in response to unilateral MCAO. The fact that NRP1/Fer kinase/CRMP-2 co-localize in membrane rafts early during ischemic injury suggests that the membrane rafts may form a scaffold to support and initiate NRP1/Fer/CRMP-2-mediated signal transduction in neuronal damage response during ischemia-reperfusion. Further understanding of the time-specific and membrane domain-specific protein-protein interaction may lead to the identification of therapeutic targets for stroke.
Collapse
|
46
|
Spatiotemporal control of cyclic AMP immunomodulation through the PKA-Csk inhibitory pathway is achieved by anchoring to an Ezrin-EBP50-PAG scaffold in effector T cells. FEBS Lett 2010; 584:2681-8. [PMID: 20420835 DOI: 10.1016/j.febslet.2010.04.056] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2010] [Revised: 04/16/2010] [Accepted: 04/20/2010] [Indexed: 11/23/2022]
Abstract
A variety of immunoregulatory signals to effector T cells from monocytes, macrophages and regulatory T cells act through cyclic adenosine monophosphate. In the effector T cell, the protein kinase A (PKA) type I isoenzyme localizes to lipid rafts during T cell activation and modulates directly the proximal events that take place after engagement of the T cell receptor. The most proximal target for PKA phosphorylation is C-terminal Src kinase (Csk), which initiates a negative signal pathway that fine-tunes the T cell activation process. The A kinase anchoring protein Ezrin colocalizes PKA and Csk by forming a supramolecular signaling complex consisting of PKA, Ezrin, Ezrin/radixin/moesin (ERM) binding protein of 50 kDa (EBP50), phosphoprotein associated with glycosphingolipid-enriched membrane microdomains (GEMs) (PAG) and Csk.
Collapse
|
47
|
Abstract
T cell activation begins with the interaction between an antigen-specific T cell and an antigen-presenting cell (APC). This interaction results in the formation of the immunological synapse, which had been considered to be responsible for antigen recognition and T cell activation. Recent advances in imaging analysis have provided new insights into T cell activation. The T cell receptor (TCR) microclusters, TCRs, kinases, and adaptors are generated upon antigen recognition at the interfaces between the T cells and the APCs and serve as a fundamental signaling unit for T cell activation. CD28-mediated costimulation is also found to be regulated by the formation of microclusters. Therefore, the dynamic regulations of TCR and CD28 microcluster formation, migration, and interaction are the key events for the initiation of T cell-mediated immune responses. Comprehensive analyses of the composition and characteristics of the TCR microcluster have identified its dynamic features. This review will outline new discoveries of the microclusters and its related concept in T cell activation.
Collapse
|
48
|
The adaptor protein EBP50 is important for localization of the protein kinase A-Ezrin complex in T-cells and the immunomodulating effect of cAMP. Biochem J 2009; 425:381-8. [PMID: 19857202 DOI: 10.1042/bj20091136] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
We recently reported that the dual-specificity AKAP (A-kinaseanchoring protein) Ezrin targets type I PKA (protein kinase A) to the vicinity of the TCR (T-cell receptor) in T-cells and, together with PAG (phosphoprotein associated with glycosphingolipid-enriched membrane microdomains) and EBP50 [ERM (Ezrin/Radixin/Moesin)-binding phosphoprotein 50], forms a scaffold that positions PKA close to its substrate, Csk (C-terminal Src kinase). This complex is important for controlling the activation state of T-cells. Ezrin binds the adaptor protein EBP50, which again contacts PAG. In the present study, we show that Ezrin and EBP50 interact with high affinity (KD=58+/-7 nM). A peptide corresponding to the EB (Ezrin-binding) region in EBP50 (EBP50pep) was used to further characterize the binding kinetics and compete the Ezrin-EBP50 interaction by various methods in vitro. Importantly, loading T-cells with EBP50pep delocalized Ezrin, but not EBP50. Furthermore, disruption of this complex interfered with cAMP modulation of T-cell activation, which is seen as a reversal of cAMP-mediated inhibition of IL-2 (interleukin 2) production, demonstrating an important role of EBP50 in this complex. In summary, both the biochemical and functional data indicate that targeting the Ezrin-EBP interaction could be a novel and potent strategy for immunomodulation.
Collapse
|
49
|
Chen Y, Veracini L, Benistant C, Jacobson K. The transmembrane protein CBP plays a role in transiently anchoring small clusters of Thy-1, a GPI-anchored protein, to the cytoskeleton. J Cell Sci 2009; 122:3966-72. [PMID: 19825940 DOI: 10.1242/jcs.049346] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It remains unclear how GPI-anchored proteins (GPIAPs), which lack cytoplasmic domains, transduce signals triggered by specific ligation. Such signal transduction has been speculated to require the ligated GPIAP to associate with membrane-spanning proteins that communicate with obligate cytoplasmic proteins. Transient anchorage of crosslinked proteins on the cell surface was previously characterized by single-particle tracking, and temporary association with the actin cytoskeleton was hypothesized to cause regulated anchorage. GPIAPs, such as Thy-1, require clustering, cholesterol and Src-family kinase (SFK) activity to become transiently anchored. By contrast, a transmembrane protein, the cystic fibrosis transmembrane conductance regulator (CFTR), which has a PDZ-binding motif in its cytoplasmic C-terminus that binds the ERM adaptor EBP50, exhibits anchorage that strictly requires EBP50 but has little dependence on cholesterol or SFK. We hypothesized that a transmembrane protein would be required to mediate the linkage between Thy-1 and the cytoskeleton. Here, we present evidence, obtained by shRNA knockdown, that the transmembrane protein Csk-binding protein (CBP) plays an obligatory role in the transient anchorage of Thy1. Furthermore, either a dominant-negative form of CBP that did not bind EBP50 or a dominant-negative EBP50 drastically reduced transient anchorage of Thy-1, indicating the involvement of this adaptor. Finally, we speculate on the role of phosphorylation in the regulation of transient anchorage.
Collapse
Affiliation(s)
- Yun Chen
- Department of Cell and Developmental Biology, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7090, USA
| | | | | | | |
Collapse
|
50
|
Feng X, Lu X, Man X, Zhou W, Jiang LQ, Knyazev P, Lei L, Huang Q, Ullrich A, Zhang Z, Chen Z. Overexpression of Csk-binding protein contributes to renal cell carcinogenesis. Oncogene 2009; 28:3320-31. [DOI: 10.1038/onc.2009.185] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|