1
|
Zhang Y, Ren Y, Zhou T, Qian Z, Bao Z. Vav family exchange factors: Potential regulator in atherosclerosis. Biochem Biophys Rep 2024; 40:101878. [PMID: 39649800 PMCID: PMC11625217 DOI: 10.1016/j.bbrep.2024.101878] [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: 08/02/2024] [Revised: 10/26/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024] Open
Abstract
The Vav family of guanosine nucleotide exchange factors (GEFs) regulates the phosphorylation of tyrosinase, influencing various physiological and pathological processes by modulating the binding of Rho GTPases to GDP/GTP. Recent research has highlighted the critical role of Vav family activation in tumorigenesis, neurological disorders, immune-related dysfunctions, and other diseases. This review offers a comprehensive overview of the structure and function of Vav proteins and their significant impact on the pathophysiology of atherosclerosis. In addition, we pay attention to the development of diagnostic and therapeutic targets centered around Vav proteins.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214002, China
| | - Yongwei Ren
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, China
| | - Tao Zhou
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, China
| | - Zhengtao Qian
- Department of Clinical Laboratory, Changshu Medicine Examination Institute, Changshu, 215500, China
| | - Zhengyang Bao
- Department of Internal Medicine, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, China
| |
Collapse
|
2
|
Kaminski S, Adjali O, Jacquet C, Garaude J, Keriel A, Lassaux A, Hipskind R, Sitbon M, Taylor N, Villalba M. The protooncogene Vav1 regulates murine leukemia virus-induced T-cell leukemogenesis. Oncoimmunology 2021; 1:600-608. [PMID: 22934252 PMCID: PMC3429564 DOI: 10.4161/onci.20225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Vav1 is expressed exclusively in hematopoietic cells and is required for T cell development and activation. Vav1-deficient mice show thymic hypocellularity due to a partial block during thymocyte development at the DN3 stage and between the double positive (DP) and single positive (SP) transition. Vav1 has been shown to play a significant role in several non-hematopoietic tumors but its role in leukemogenesis is unknown. To address this question, we investigated the role of Vav1 in retrovirus-induced T cell leukemogenesis. Infection of Vav1-deficient mice with the Moloney strain of murine leukemia virus (M-MuLV) significantly affected tumor phenotype without modulating tumor incidence or latency. M-MuLV-infected Vav1-deficient mice showed reduced splenomegaly, higher hematocrit levels and hypertrophic thymi. Notably, Vav1-deficient mice with M-MuLV leukemias presented with markedly lower TCRβ/CD3 levels, indicating that transformation occurred at an earlier stage of T cell development than in WT mice. Thus, impaired T cell development modulates the outcome of retrovirus-induced T cell leukemias, demonstrating a link between T cell development and T cell leukemogenesis.
Collapse
Affiliation(s)
- Sandra Kaminski
- Institut de Génétique Moléculaire de Montpellier; UMR 5535; CNRS; Montpellier, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Sadegh-Nasseri S. How a Proposed Hypothesis during My PhD Training Shaped My Career. Crit Rev Immunol 2020; 40:449-464. [PMID: 33463956 PMCID: PMC11014643 DOI: 10.1615/critrevimmunol.2020035324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In this memoir-style essay, I have narrated the evolution of my scientific career, as deeply influenced by my PhD training and the mentorship of Professor Eli Sercarz. Starting in his lab, and continuing to my own laboratory, many of the questions we have pursued link in some way to Eli's ideas. In this essay, I have summarized the path that I followed after graduating from his lab and highlight findings along the way. I apologize to my colleagues whose work was not discussed here due to the nature of this review and space limitations.
Collapse
|
4
|
Wang B, Zhang W, Jankovic V, Golubov J, Poon P, Oswald EM, Gurer C, Wei J, Ramos I, Wu Q, Waite J, Ni M, Adler C, Wei Y, Macdonald L, Rowlands T, Brydges S, Siao J, Poueymirou W, MacDonald D, Yancopoulos GD, Sleeman MA, Murphy AJ, Skokos D. Combination cancer immunotherapy targeting PD-1 and GITR can rescue CD8+ T cell dysfunction and maintain memory phenotype. Sci Immunol 2019; 3:3/29/eaat7061. [PMID: 30389797 DOI: 10.1126/sciimmunol.aat7061] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/11/2018] [Indexed: 12/13/2022]
Abstract
Most patients with cancer do not develop durable antitumor responses after programmed cell death protein 1 (PD-1) or programmed cell death ligand 1(PD-L1) checkpoint inhibition monotherapy because of an ephemeral reversal of T cell dysfunction and failure to promote long-lasting immunological T cell memory. Activating costimulatory pathways to induce stronger T cell activation may improve the efficacy of checkpoint inhibition and lead to durable antitumor responses. We performed single-cell RNA sequencing of more than 2000 tumor-infiltrating CD8+ T cells in mice receiving both PD-1 and GITR (glucocorticoid-induced tumor necrosis factor receptor-related protein) antibodies and found that this combination synergistically enhanced the effector function of expanded CD8+ T cells by restoring the balance of key homeostatic regulators CD226 and T cell immunoreceptor with Ig and ITIM domains (TIGIT), leading to a robust survival benefit. Combination therapy decreased CD8+ T cell dysfunction and induced a highly proliferative precursor effector memory T cell phenotype in a CD226-dependent manner. PD-1 inhibition rescued CD226 activity by preventing PD-1-Src homology region 2 (SHP2) dephosphophorylation of the CD226 intracellular domain, whereas GITR agonism decreased TIGIT expression. Unmasking the molecular pathways driving durable antitumor responses will be essential to the development of rational approaches to optimizing cancer immunotherapy.
Collapse
Affiliation(s)
- Bei Wang
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | - Wen Zhang
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | | | | | - Patrick Poon
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | - Erin M Oswald
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | - Cagan Gurer
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | - Joyce Wei
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | - Ilyssa Ramos
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | - Qi Wu
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | - Janelle Waite
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | - Min Ni
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | - Christina Adler
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | - Yi Wei
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | - Lynn Macdonald
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | - Tracey Rowlands
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | | | - Jean Siao
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | | | | | | | | | - Andrew J Murphy
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA
| | - Dimitris Skokos
- Regeneron Pharmaceuticals,Tarrytown, New York, NY 10591, USA.
| |
Collapse
|
5
|
Bedi S, Ono A. Friend or Foe: The Role of the Cytoskeleton in Influenza A Virus Assembly. Viruses 2019; 11:v11010046. [PMID: 30634554 PMCID: PMC6356976 DOI: 10.3390/v11010046] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/02/2019] [Accepted: 01/08/2019] [Indexed: 12/12/2022] Open
Abstract
Influenza A Virus (IAV) is a respiratory virus that causes seasonal outbreaks annually and pandemics occasionally. The main targets of the virus are epithelial cells in the respiratory tract. Like many other viruses, IAV employs the host cell’s machinery to enter cells, synthesize new genomes and viral proteins, and assemble new virus particles. The cytoskeletal system is a major cellular machinery, which IAV exploits for its entry to and exit from the cell. However, in some cases, the cytoskeleton has a negative impact on efficient IAV growth. In this review, we highlight the role of cytoskeletal elements in cellular processes that are utilized by IAV in the host cell. We further provide an in-depth summary of the current literature on the roles the cytoskeleton plays in regulating specific steps during the assembly of progeny IAV particles.
Collapse
Affiliation(s)
- Sukhmani Bedi
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Akira Ono
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
6
|
Dekraker C, Boucher E, Mandato CA. Regulation and Assembly of Actomyosin Contractile Rings in Cytokinesis and Cell Repair. Anat Rec (Hoboken) 2018; 301:2051-2066. [PMID: 30312008 DOI: 10.1002/ar.23962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 08/24/2018] [Accepted: 08/27/2018] [Indexed: 01/17/2023]
Abstract
Cytokinesis and single-cell wound repair both involve contractile assemblies of filamentous actin (F-actin) and myosin II organized into characteristic ring-like arrays. The assembly of these actomyosin contractile rings (CRs) is specified spatially and temporally by small Rho GTPases, which trigger local actin polymerization and myosin II contractility via a variety of downstream effectors. We now have a much clearer view of the Rho GTPase signaling cascade that leads to the formation of CRs, but some factors involved in CR positioning, assembly, and function remain poorly understood. Recent studies show that this regulation is multifactorial and goes beyond the long-established Ca2+ -dependent processes. There is substantial evidence that the Ca2+ -independent changes in cell shape, tension, and plasma membrane composition that characterize cytokinesis and single-cell wound repair also regulate CR formation. Elucidating the regulation and mechanistic properties of CRs is important to our understanding of basic cell biology and holds potential for therapeutic applications in human disease. In this review, we present a primer on the factors influencing and regulating CR positioning, assembly, and contraction as they occur in a variety of cytokinetic and single-cell wound repair models. Anat Rec, 301:2051-2066, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Corina Dekraker
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Eric Boucher
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Craig A Mandato
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Song X, Gong M, Chen Y, Liu H, Zhang J. Nine hub genes as the potential indicator for the clinical outcome of diabetic nephropathy. J Cell Physiol 2018; 234:1461-1468. [PMID: 30078220 DOI: 10.1002/jcp.26958] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 06/13/2018] [Indexed: 01/08/2023]
Abstract
Diabetic nephropathy (DN) is one of the most serious and dangerous chronic complications caused by diabetes mellitus, and the identification and development of novel biomarkers could be beneficial for the diagnosis and prognosis of DN patients. This study focused on researching the differently expressed pattern of the DN samples from glomeruli and tubulointerstitium. Significance analysis of microarrays (SAM) was used to identify differentially regulated genes in 44 microdissected human kidney samples. Functional enrichment analysis was used to analyze the functions these genes are mostly enriched in. Besides, protein-protein interaction (PPI) network was used to select the hub genes that were associated with DN. The gene expression pattern of DN samples from glomeruli and tubulointerstitium was found to be quite different. It showed that the recurrence rate of DN originating from glomeruli and control samples was lower than that from tubulointerstitium and control samples. A total of 332 differentially expressed genes were identified between glomeruli tissues and tubulointerstitium tissues. PPI network analysis was performed on these 332 genes. Finally, nine hub genes were selected as the most potential biomarkers in the occurrence of DN. In conclusion, a total of 332 genes were found to be related to DN, and these genes were found to be mostly enriched in pathways such as immunity, inflammatory, and vascular pathways. Three genes VAV1, LCK, and Plk had the potential to serve as indicators for the occurrence and development of DN in clinical management.
Collapse
Affiliation(s)
- Xiaoping Song
- Department of Laboratory, Qingdao Hiser Medical Group, Qingdao, China
| | - Min Gong
- Department of Endocrinology, Qingdao Hiser Medical Group, Qingdao, China
| | - Yanping Chen
- Department of Laboratory, Qingdao Women and Children's Hospital, Qingdao, China
| | - Hui Liu
- Department of Laboratory, Qingdao Women and Children's Hospital, Qingdao, China
| | - Jun Zhang
- Department of Emergency, Qingdao Municipal Hospital East Hospital, Qingdao, China
| |
Collapse
|
8
|
Gaud G, Roncagalli R, Chaoui K, Bernard I, Familiades J, Colacios C, Kassem S, Monsarrat B, Burlet-Schiltz O, de Peredo AG, Malissen B, Saoudi A. The costimulatory molecule CD226 signals through VAV1 to amplify TCR signals and promote IL-17 production by CD4 + T cells. Sci Signal 2018; 11:11/538/eaar3083. [PMID: 29991650 DOI: 10.1126/scisignal.aar3083] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The activation of T cells requires the guanine nucleotide exchange factor VAV1. Using mice in which a tag for affinity purification was attached to endogenous VAV1 molecules, we analyzed by quantitative mass spectrometry the signaling complex that assembles around activated VAV1. Fifty VAV1-binding partners were identified, most of which had not been previously reported to participate in VAV1 signaling. Among these was CD226, a costimulatory molecule of immune cells. Engagement of CD226 induced the tyrosine phosphorylation of VAV1 and synergized with T cell receptor (TCR) signals to specifically enhance the production of interleukin-17 (IL-17) by primary human CD4+ T cells. Moreover, co-engagement of the TCR and a risk variant of CD226 that is associated with autoimmunity (rs763361) further enhanced VAV1 activation and IL-17 production. Thus, our study reveals that a VAV1-based, synergistic cross-talk exists between the TCR and CD226 during both physiological and pathological T cell responses and provides a rational basis for targeting CD226 for the management of autoimmune diseases.
Collapse
Affiliation(s)
- Guillaume Gaud
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, CNRS, Inserm, Toulouse 31300, France
| | - Romain Roncagalli
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, Inserm, CNRS, 13288 Marseille, France
| | - Karima Chaoui
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS UMR 5089, 31077 Toulouse Cedex, France
| | - Isabelle Bernard
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, CNRS, Inserm, Toulouse 31300, France
| | - Julien Familiades
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, CNRS, Inserm, Toulouse 31300, France
| | - Céline Colacios
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, CNRS, Inserm, Toulouse 31300, France
| | - Sahar Kassem
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, CNRS, Inserm, Toulouse 31300, France
| | - Bernard Monsarrat
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS UMR 5089, 31077 Toulouse Cedex, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS UMR 5089, 31077 Toulouse Cedex, France
| | - Anne Gonzalez de Peredo
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS UMR 5089, 31077 Toulouse Cedex, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, Inserm, CNRS, 13288 Marseille, France.,Centre d'Immunophénomique, Aix-Marseille Université, Inserm, CNRS, 13288 Marseille, France
| | - Abdelhadi Saoudi
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, CNRS, Inserm, Toulouse 31300, France.
| |
Collapse
|
9
|
Zumerle S, Molon B, Viola A. Membrane Rafts in T Cell Activation: A Spotlight on CD28 Costimulation. Front Immunol 2017; 8:1467. [PMID: 29163534 PMCID: PMC5675840 DOI: 10.3389/fimmu.2017.01467] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/19/2017] [Indexed: 12/28/2022] Open
Abstract
Spatiotemporal compartmentalization of signaling pathways and second messengers is pivotal for cell biology and membrane rafts are, therefore, required for several lymphocyte functions. On the other hand, T cells have the specific necessity of tuning signaling amplification depending on the context in which the antigen is presented. In this review, we discuss of membrane rafts in the context of T cell signaling, focusing on CD28-mediated costimulation.
Collapse
Affiliation(s)
- Sara Zumerle
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Barbara Molon
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Pediatric Research Institute "Citta della Speranza", Padova, Italy
| |
Collapse
|
10
|
Jung SH, Yoo EH, Yu MJ, Song HM, Kang HY, Cho JY, Lee JR. ARAP, a Novel Adaptor Protein, Is Required for TCR Signaling and Integrin-Mediated Adhesion. THE JOURNAL OF IMMUNOLOGY 2016; 197:942-52. [PMID: 27335501 DOI: 10.4049/jimmunol.1501913] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 05/19/2016] [Indexed: 11/19/2022]
Abstract
A novel adaptor protein was identified by analyzing phosphotyrosine proteomes from membrane rafts of activated T cells. This protein showed sequence similarity to a well-known T cell adaptor protein, adhesion and degranulation-promoting adaptor protein (ADAP); therefore, the novel protein was designated activation-dependent, raft-recruited ADAP-like phosphoprotein (ARAP). Suppression of ARAP impaired the major signaling pathways downstream of the TCR. ARAP associated with the Src homology 2 domain of Src homology 2-containing leukocyte protein of 76 kDa via the phosphorylation of two YDDV motifs in response to TCR stimulation. ARAP also mediated integrin activation but was not involved in actin polymerization. The results of this study indicate that a novel T cell adaptor protein, ARAP, plays a unique role in T cells as a part of both the proximal activation signaling and inside-out signaling pathways that result in integrin activation and T cell adhesion.
Collapse
Affiliation(s)
- Seung Hee Jung
- Department of Life Science, College of Natural Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Eun Hye Yoo
- Department of Life Science, College of Natural Sciences, Ewha Womans University, Seoul 03760, Korea; Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea; and
| | - Mi Jin Yu
- Department of Life Science, College of Natural Sciences, Ewha Womans University, Seoul 03760, Korea; Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea; and
| | - Hyeon Myeong Song
- Department of Life Science, College of Natural Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Hee Yoon Kang
- Department of Life Science, College of Natural Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Je-Yoel Cho
- Department of Veterinary Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Jong Ran Lee
- Department of Life Science, College of Natural Sciences, Ewha Womans University, Seoul 03760, Korea; Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea; and
| |
Collapse
|
11
|
Integrated mate-pair and RNA sequencing identifies novel, targetable gene fusions in peripheral T-cell lymphoma. Blood 2016; 128:1234-45. [PMID: 27297792 DOI: 10.1182/blood-2016-03-707141] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022] Open
Abstract
Peripheral T-cell lymphomas (PTCLs) represent a heterogeneous group of T-cell malignancies that generally demonstrate aggressive clinical behavior, often are refractory to standard therapy, and remain significantly understudied. The most common World Health Organization subtype is PTCL, not otherwise specified (NOS), essentially a "wastebasket" category because of inadequate understanding to assign cases to a more specific diagnostic entity. Identification of novel fusion genes has contributed significantly to improving the classification, biologic understanding, and therapeutic targeting of PTCLs. Here, we integrated mate-pair DNA and RNA next-generation sequencing to identify chromosomal rearrangements encoding expressed fusion transcripts in PTCL, NOS. Two of 11 cases had novel fusions involving VAV1, encoding a truncated form of the VAV1 guanine nucleotide exchange factor important in T-cell receptor signaling. Fluorescence in situ hybridization studies identified VAV1 rearrangements in 10 of 148 PTCLs (7%). These were observed exclusively in PTCL, NOS (11%) and anaplastic large cell lymphoma (11%). In vitro, ectopic expression of a VAV1 fusion promoted cell growth and migration in a RAC1-dependent manner. This growth was inhibited by azathioprine, a clinically available RAC1 inhibitor. We also identified novel kinase gene fusions, ITK-FER and IKZF2-ERBB4, as candidate therapeutic targets that show similarities to known recurrent oncogenic ITK-SYK fusions and ERBB4 transcript variants in PTCLs, respectively. Additional novel and potentially clinically relevant fusions also were discovered. Together, these findings identify VAV1 fusions as recurrent and targetable events in PTCLs and highlight the potential for clinical sequencing to guide individualized therapy approaches for this group of aggressive malignancies.
Collapse
|
12
|
Tian J, Nickels J, Katsaras J, Cheng X. Behavior of Bilayer Leaflets in Asymmetric Model Membranes: Atomistic Simulation Studies. J Phys Chem B 2016; 120:8438-48. [DOI: 10.1021/acs.jpcb.6b02148] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | - John Katsaras
- The Bredesen Center for Interdisciplinary Research and Graduate Education, 444 Greve Hall, 821 Volunteer Boulevard, Knoxville, Tennessee 37996-3394, United States
| | | |
Collapse
|
13
|
Zynda ER, Grimm MJ, Yuan M, Zhong L, Mace TA, Capitano M, Ostberg JR, Lee KP, Pralle A, Repasky EA. A role for the thermal environment in defining co-stimulation requirements for CD4(+) T cell activation. Cell Cycle 2016; 14:2340-54. [PMID: 26131730 DOI: 10.1080/15384101.2015.1049782] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Maintenance of normal core body temperature is vigorously defended by long conserved, neurovascular homeostatic mechanisms that assist in heat dissipation during prolonged, heat generating exercise or exposure to warm environments. Moreover, during febrile episodes, body temperature can be significantly elevated for at least several hours at a time. Thus, as blood cells circulate throughout the body, physiologically relevant variations in surrounding tissue temperature can occur; moreover, shifts in core temperature occur during daily circadian cycles. This study has addressed the fundamental question of whether the threshold of stimulation needed to activate lymphocytes is influenced by temperature increases associated with physiologically relevant increases in temperature. We report that the need for co-stimulation of CD4+ T cells via CD28 ligation for the production of IL-2 is significantly reduced when cells are exposed to fever-range temperature. Moreover, even in the presence of sufficient CD28 ligation, provision of extra heat further increases IL-2 production. Additional in vivo and in vitro data (using both thermal and chemical modulation of membrane fluidity) support the hypothesis that the mechanism by which temperature modulates co-stimulation is linked to increases in membrane fluidity and membrane macromolecular clustering in the plasma membrane. Thermally-regulated changes in plasma membrane organization in response to physiological increases in temperature may assist in the geographical control of lymphocyte activation, i.e., stimulating activation in lymph nodes rather than in cooler surface regions, and further, may temporarily and reversibly enable CD4+ T cells to become more quickly and easily activated during times of infection during fever.
Collapse
Key Words
- APC, antigen-presenting cell
- CD28, cluster of differentiation 28
- CD3, cluster of differentiation 3
- CD4, cluster of differentiation 4
- CD8, cluster of differentiation 8
- CTLA-4, cytotoxic T-lymphocyte-associated protein 4
- CTxB, cholera toxin B subunit
- Ct, cycle threshold
- ELISA, enzyme-linked immunosorbant assay
- EtOH, ethanol
- FITC, fluoroisothiocyanate
- GM1, monosialotetrahexosylganglioside
- IDEAS, imagestream data exploration and analysis software
- IL-2, interleukin 2
- LA, latrunculin A
- MβCD, methyl-β-cyclodextrin
- PD-1, Programmed cell death-1
- PMA, phorbol 12-myristate 13-acetate
- T cell activation
- T cell co-stimulation
- TCR, T cell receptor
- TDI, time delay integration
- TMA-DPH, trimethylammonium diphenylhexatriene
- WBH, whole body hyperthermia.
- fever
- hyperthermia
- immune response
- membrane fluidity
- pMHC, peptide-major histocompatibility complexes
- qRT-PCR, quantitative reverse transcription polymerase chain reaction
Collapse
Affiliation(s)
- Evan R Zynda
- a Department of Cell Stress Biology ; Roswell Park Cancer Institute ; Buffalo , NY USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Saslowsky DE, Thiagarajah JR, McCormick BA, Lee JC, Lencer WI. Microbial sphingomyelinase induces RhoA-mediated reorganization of the apical brush border membrane and is protective against invasion. Mol Biol Cell 2016; 27:1120-30. [PMID: 26864627 PMCID: PMC4814219 DOI: 10.1091/mbc.e15-05-0293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 02/01/2016] [Indexed: 12/19/2022] Open
Abstract
Both commensal and pathogenic microbes that colonize the GI tract can synthesize and secrete spingomyelinase enzymes that cleave membrane sphingomyelin, leaving the ceramide component intact in the cell membrane. This study examines how this reaction affects the structure and function of host enterocytes and mucosal defense. The apical brush border membrane (BBM) of intestinal epithelial cells forms a highly structured and dynamic environmental interface that serves to regulate cellular physiology and block invasion by intestinal microbes and their products. How the BBM dynamically responds to pathogenic and commensal bacterial signals can define intestinal homeostasis and immune function. We previously found that in model intestinal epithelium, the conversion of apical membrane sphingomyelin to ceramide by exogenous bacterial sphingomyelinase (SMase) protected against the endocytosis and toxicity of cholera toxin. Here we elucidate a mechanism of action by showing that SMase induces a dramatic, reversible, RhoA-dependent alteration of the apical cortical F-actin network. Accumulation of apical membrane ceramide is necessary and sufficient to induce the actin phenotype, and this coincides with altered membrane structure and augmented innate immune function as evidenced by resistance to invasion by Salmonella.
Collapse
Affiliation(s)
- David E Saslowsky
- Division of Gastroenterology and Nutrition, Boston Children's Hospital, Boston, MA 02115 Harvard Digestive Diseases Center, Boston Children's Hospital, Boston, MA 02115 Harvard Medical School, Boston, MA 02115
| | - Jay R Thiagarajah
- Division of Gastroenterology and Nutrition, Boston Children's Hospital, Boston, MA 02115 Harvard Digestive Diseases Center, Boston Children's Hospital, Boston, MA 02115 Harvard Medical School, Boston, MA 02115
| | - Beth A McCormick
- Harvard Digestive Diseases Center, Boston Children's Hospital, Boston, MA 02115 Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655
| | - Jean C Lee
- Harvard Medical School, Boston, MA 02115 Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115
| | - Wayne I Lencer
- Division of Gastroenterology and Nutrition, Boston Children's Hospital, Boston, MA 02115 Harvard Digestive Diseases Center, Boston Children's Hospital, Boston, MA 02115 Harvard Medical School, Boston, MA 02115
| |
Collapse
|
15
|
Nickels JD, Smith JC, Cheng X. Lateral organization, bilayer asymmetry, and inter-leaflet coupling of biological membranes. Chem Phys Lipids 2015; 192:87-99. [DOI: 10.1016/j.chemphyslip.2015.07.012] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/20/2015] [Accepted: 07/25/2015] [Indexed: 11/28/2022]
|
16
|
Côte M, Fos C, Canonigo-Balancio AJ, Ley K, Bécart S, Altman A. SLAT promotes TCR-mediated, Rap1-dependent LFA-1 activation and adhesion through interaction of its PH domain with Rap1. J Cell Sci 2015; 128:4341-52. [PMID: 26483383 DOI: 10.1242/jcs.172742] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 10/09/2015] [Indexed: 01/13/2023] Open
Abstract
SLAT (also known as DEF6) promotes T cell activation and differentiation by regulating NFAT-Ca(2+) signaling. However, its role in TCR-mediated inside-out signaling, which induces integrin activation and T cell adhesion, a central process in T cell immunity and inflammation, has not been explored. Here, we show that SLAT is crucial for TCR-induced adhesion to ICAM-1 and affinity maturation of LFA-1 in CD4(+) T cells. Mechanistic studies revealed that SLAT interacts, through its PH domain, with a key component of inside-out signaling, namely the active form of the small GTPase Rap1 (which has two isoforms, Rap1A and Rap1B). This interaction has been further shown to facilitate the interdependent recruitment of Rap1 and SLAT to the T cell immunological synapse upon TCR engagement. Furthermore, a SLAT mutant lacking its PH domain drastically inhibited LFA-1 activation and CD4(+) T cell adhesion. Finally, we established that a constitutively active form of Rap1, which is present at the plasma membrane, rescues the defective LFA-1 activation and ICAM-1 adhesion in SLAT-deficient (Def6(-/-)) T cells. These findings ascribe a new function to SLAT, and identify Rap1 as a target of SLAT function in TCR-mediated inside-out signaling.
Collapse
Affiliation(s)
- Marjorie Côte
- Division of Cell Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Camille Fos
- Division of Cell Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Ann J Canonigo-Balancio
- Division of Cell Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Stéphane Bécart
- Division of Cell Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Amnon Altman
- Division of Cell Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| |
Collapse
|
17
|
Cerecedo D, Martínez‐Vieyra I, Maldonado‐García D, Hernández‐González E, Winder SJ. Association of Membrane/Lipid Rafts With the Platelet Cytoskeleton and the Caveolin PY14: Participation in the Adhesion Process. J Cell Biochem 2015; 116:2528-40. [DOI: 10.1002/jcb.25197] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 04/14/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Doris Cerecedo
- Laboratorio de HematobiologíaEscuela Nacional de Medicina y Homeopatía (ENMH)Instituto Politécnico Nacional (IPN)Mexico CityMexico
| | - Ivette Martínez‐Vieyra
- Laboratorio de HematobiologíaEscuela Nacional de Medicina y Homeopatía (ENMH)Instituto Politécnico Nacional (IPN)Mexico CityMexico
| | - Deneb Maldonado‐García
- Departamento de Biología CelularCentro de Investigación y de Estudios Avanzados del IPN (Cinvestav‐IPN)Mexico CityMexico
| | - Enrique Hernández‐González
- Departamento de Biología CelularCentro de Investigación y de Estudios Avanzados del IPN (Cinvestav‐IPN)Mexico CityMexico
| | - Steve J. Winder
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| |
Collapse
|
18
|
Helou YA, Petrashen AP, Salomon AR. Vav1 Regulates T-Cell Activation through a Feedback Mechanism and Crosstalk between the T-Cell Receptor and CD28. J Proteome Res 2015; 14:2963-75. [PMID: 26043137 DOI: 10.1021/acs.jproteome.5b00340] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Vav1, a Rac/Rho guanine nucleotide exchange factor and a critical component of the T-cell receptor (TCR) signaling cascade is tyrosine phosphorylated rapidly in response to T-cell activation. Vav1 has established roles in proliferation, cytokine secretion, Ca(2+) responses, and actin cytoskeleton regulation; however, its function in the regulation of phosphorylation of TCR components, including the ζ chain, the CD3 δ, ε, γ chains, and the associated kinases Lck and ZAP-70, is not well established. To obtain a more comprehensive picture of the role of Vav1 in receptor proximal signaling, we performed a wide-scale characterization of Vav1-dependent tyrosine phosphorylation events using quantitative phosphoproteomic analysis of Vav1-deficient T cells across a time course of TCR stimulation. Importantly, this study revealed a new function for Vav1 in the negative feedback regulation of the phosphorylation of immunoreceptor tyrosine-based activation motifs within the ζ chains, CD3 δ, ε, γ chains, as well as activation sites on the critical T cell tyrosine kinases Itk, Lck, and ZAP-70. Our study also uncovered a previously unappreciated role for Vav1 in crosstalk between the CD28 and TCR signaling pathways.
Collapse
Affiliation(s)
- Ynes A Helou
- †Department of Molecular Pharmacology, Physiology, and Biotechnology and ‡Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02903, United States
| | - Anna P Petrashen
- †Department of Molecular Pharmacology, Physiology, and Biotechnology and ‡Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02903, United States
| | - Arthur R Salomon
- †Department of Molecular Pharmacology, Physiology, and Biotechnology and ‡Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02903, United States
| |
Collapse
|
19
|
Xu L, Auzins A, Sun X, Xu Y, Harnischfeger F, Lu Y, Li Z, Chen YH, Zheng W, Liu W. The synaptic recruitment of lipid rafts is dependent on CD19-PI3K module and cytoskeleton remodeling molecules. J Leukoc Biol 2015; 98:223-34. [PMID: 25979433 DOI: 10.1189/jlb.2a0614-287rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 04/15/2015] [Indexed: 11/24/2022] Open
Abstract
Sphingolipid- and cholesterol-rich lipid raft microdomains are important in the initiation of BCR signaling. Although it is known that lipid rafts promote the coclustering of BCR and Lyn kinase microclusters within the B cell IS, the molecular mechanism of the recruitment of lipid rafts into the B cell IS is not understood completely. Here, we report that the synaptic recruitment of lipid rafts is dependent on the cytoskeleton-remodeling proteins, RhoA and Vav. Such an event is also efficiently regulated by motor proteins, myosin IIA and dynein. Further evidence suggests the synaptic recruitment of lipid rafts is, by principle, an event triggered by BCR signaling molecules and second messenger molecules. BCR-activating coreceptor CD19 potently enhances such an event depending on its cytoplasmic Tyr421 and Tyr482 residues. The enhancing function of the CD19-PI3K module in synaptic recruitment of lipid rafts is also confirmed in human peripheral blood B cells. Thus, these results improve our understanding of the molecular mechanism of the recruitment of lipid raft microdomains in B cell IS.
Collapse
Affiliation(s)
- Liling Xu
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Arturs Auzins
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaolin Sun
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yinsheng Xu
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Fiona Harnischfeger
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yun Lu
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Zhanguo Li
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Ying-Hua Chen
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Wenjie Zheng
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Wanli Liu
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| |
Collapse
|
20
|
Chen Q, Zhang M, Li Y, Xu D, Wang Y, Song A, Zhu B, Huang Y, Zheng JC. CXCR7 Mediates Neural Progenitor Cells Migration to CXCL12 Independent of CXCR4. Stem Cells 2015; 33:2574-85. [PMID: 25833331 DOI: 10.1002/stem.2022] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 03/02/2015] [Indexed: 12/26/2022]
Abstract
Neural progenitor cell (NPC) migration is an essential process for brain development, adult neurogenesis, and neuroregeneration after brain injury. Stromal cell-derived factor-1 (SDF-1, CXCL12) and its traditional receptor CXCR4 are well known to regulate NPC migration. However, the discovery of CXCR7, a newly identified CXCL12 receptor, adds to the dynamics of the existing CXCL12/CXCR4 pair. Antagonists for either CXCR4 or CXCR7 blocked CXCL12-mediated NPC migration in a transwell chemotaxis assay, suggesting that both receptors are required for CXCL12 action. We derived NPC cultures from Cxcr4 knockout (KO) mice and used transwell and stripe assays to determine the cell migration. NPCs derived from Cxcr4 KO mice polarized and migrated in response to CXCL12 gradient, suggesting that CXCR7 could serve as an independent migration receptor. Furthermore, Cxcr4 KO NPCs transplanted into the adult mouse striatum migrated in response to the adjacent injection of CXCL12, an effect that was blocked by a CXCR7 antagonist, suggesting that CXCR7 also mediates NPC migration in vivo. Molecular mechanism studies revealed that CXCR7 interact with Rac1 in the leading edge of the polarized NPCs in the absence of CXCR4. Both CXCR7 and Rac1 are required for extracellular signal-regulated kinases (ERK) 1/2 activation and subsequent NPC migration, indicating that CXCR7 could serve as a functional receptor in CXCL12-mediated NPC migration independent of CXCR4. Together these results reveal an essential role of CXCR7 for CXCL12-mediated NPC migration that will be important to understand neurogenesis during development and in adulthood.
Collapse
Affiliation(s)
- Qiang Chen
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Min Zhang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yuju Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Dongsheng Xu
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Yi Wang
- Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Aihong Song
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Bing Zhu
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Yunlong Huang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jialin C Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
21
|
Saoudi A, Kassem S, Dejean A, Gaud G. Rho-GTPases as key regulators of T lymphocyte biology. Small GTPases 2014; 5:28208. [PMID: 24825161 DOI: 10.4161/sgtp.28208] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Rho-GTPases belong to the Ras superfamily and are crucial signal transducing proteins downstream of many receptors. In general, the Rho-GTPases function as molecular switches, cycling between inactive (GDP-bound) and active (GTP-bound) states. The activated GTP bound Rho-GTPases interact with a broad spectrum of effectors to regulate a plethora of biological pathways including cytoskeletal dynamics, motility, cytokinesis, cell growth, apoptosis, transcriptional activity and nuclear signaling. Recently, gene targeting in mice allowed the selective inactivation of different Rho-GTPases and has advanced our understanding of the physiological role of these proteins, particularly in the immune system. Particularly, these proteins are key signaling molecules in T lymphocytes, which are generated in the thymus and are major players in the immune system. The scope of this review is to discuss recent data obtained in Rho-GTPases deficient mice by focusing on the role-played by Rho-GTPases in T-lymphocyte development, migration, activation and differentiation.
Collapse
Affiliation(s)
- Abdelhadi Saoudi
- Inserm; U1043; Toulouse, France; CNRS; U5282; Toulouse, France; Université de Toulouse; Centre de Physiopathologie de Toulouse Purpan; Toulouse, France
| | - Sahar Kassem
- Inserm; U1043; Toulouse, France; CNRS; U5282; Toulouse, France; Université de Toulouse; Centre de Physiopathologie de Toulouse Purpan; Toulouse, France
| | - Anne Dejean
- Inserm; U1043; Toulouse, France; CNRS; U5282; Toulouse, France; Université de Toulouse; Centre de Physiopathologie de Toulouse Purpan; Toulouse, France
| | - Guillaume Gaud
- Inserm; U1043; Toulouse, France; CNRS; U5282; Toulouse, France; Université de Toulouse; Centre de Physiopathologie de Toulouse Purpan; Toulouse, France
| |
Collapse
|
22
|
Serrano-Pertierra E, Cernuda-Morollón E, Brdička T, Hoøejši V, López-Larrea C. L-plastin is involved in NKG2D recruitment into lipid rafts and NKG2D-mediated NK cell migration. J Leukoc Biol 2014; 96:437-45. [PMID: 24803550 DOI: 10.1189/jlb.2a1013-564r] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Membrane rafts are microdomains of the plasma membrane that have multiple biological functions. The involvement of these structures in the biology of T cells, namely in signal transduction by the TCR, has been widely studied. However, the role of membrane rafts in immunoreceptor signaling in NK cells is less well known. We studied the distribution of the activating NKG2D receptor in lipid rafts by isolating DRMs in a sucrose density gradient or by raft fractionation by β-OG-selective solubility in the NKL cell line. We found that the NKG2D-DAP10 complex and pVav are recruited into rafts upon receptor stimulation. Qualitative proteomic analysis of these fractions showed that the actin cytoskeleton is involved in this process. In particular, we found that the actin-bundling protein L-plastin plays an important role in the clustering of NKG2D into lipid rafts. Moreover, coengagement of the inhibitory receptor NKG2A partially disrupted NKG2D recruitment into rafts. Furthermore, we demonstrated that L-plastin participates in NKG2D-mediated inhibition of NK cell chemotaxis.
Collapse
Affiliation(s)
| | - Eva Cernuda-Morollón
- Neurology Departments, Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - Tomáš Brdička
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic; and
| | - Václav Hoøejši
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic; and
| | | |
Collapse
|
23
|
Gudheti MV, Curthoys NM, Gould TJ, Kim D, Gunewardene MS, Gabor KA, Gosse JA, Kim CH, Zimmerberg J, Hess ST. Actin mediates the nanoscale membrane organization of the clustered membrane protein influenza hemagglutinin. Biophys J 2013; 104:2182-92. [PMID: 23708358 DOI: 10.1016/j.bpj.2013.03.054] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 03/15/2013] [Accepted: 03/20/2013] [Indexed: 12/22/2022] Open
Abstract
The influenza viral membrane protein hemagglutinin (HA) is required at high concentrations on virion and host-cell membranes for infectivity. Because the role of actin in membrane organization is not completely understood, we quantified the relationship between HA and host-cell actin at the nanoscale. Results obtained using superresolution fluorescence photoactivation localization microscopy (FPALM) in nonpolarized cells show that HA clusters colocalize with actin-rich membrane regions (ARMRs). Individual molecular trajectories in live cells indicate restricted HA mobility in ARMRs, and actin disruption caused specific changes to HA clustering. Surprisingly, the actin-binding protein cofilin was excluded from some regions within several hundred nanometers of HA clusters, suggesting that HA clusters or adjacent proteins within the same clusters influence local actin structure. Thus, with the use of imaging, we demonstrate a dynamic relationship between glycoprotein membrane organization and the actin cytoskeleton at the nanoscale.
Collapse
Affiliation(s)
- Manasa V Gudheti
- Department of Physics and Astronomy, University of Maine, Orono, ME, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Takahashi D, Kawamura Y, Uemura M. Changes of detergent-resistant plasma membrane proteins in oat and rye during cold acclimation: association with differential freezing tolerance. J Proteome Res 2013; 12:4998-5011. [PMID: 24111712 DOI: 10.1021/pr400750g] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cold acclimation (CA) results in an increase in freezing tolerance of plants, which is closely associated to functional changes of the plasma membrane (PM). Although proteomic studies have revealed compositional changes of the PM during CA, there has been no large-scale study of how the microdomains in the PM, which contains specific lipids and proteins, change during CA. Therefore, we conducted semiquantitative shotgun proteomics using microdomain-enriched detergent-resistant membrane (DRM) fractions extracted from low freezing-tolerant oat and highly freezing-tolerant rye. We identified 740 and 809 DRM proteins in oat and rye, respectively. Among the proteins identified, the abundances of a variety of proteins, such as P-type ATPase and aquaporins, were affected by CA in both oat and rye. Some CA-responsive proteins in the DRM fractions, such as heat shock protein 70, changed differently in oat and rye. In addition, changes in lipocalins and sugar transporters in the DRM fractions were different from those found in total PM fraction during CA. This is the first report to describe compositional changes in the DRM during CA. The proteomic profiles obtained in the present study hint at many possible microdomain functions associated with CA and freezing tolerance.
Collapse
Affiliation(s)
- Daisuke Takahashi
- United Graduate School of Agricultural Sciences and ‡Cryobiofrontier Research Center, Iwate University , 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | | | | |
Collapse
|
25
|
Kyriakakis E, Maslova K, Frachet A, Ferri N, Contini A, Pfaff D, Erne P, Resink TJ, Philippova M. Cross-talk between EGFR and T-cadherin: EGFR activation promotes T-cadherin localization to intercellular contacts. Cell Signal 2013; 25:1044-53. [DOI: 10.1016/j.cellsig.2013.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 01/17/2013] [Accepted: 02/05/2013] [Indexed: 01/09/2023]
|
26
|
Kong KF, Altman A. In and out of the bull's eye: protein kinase Cs in the immunological synapse. Trends Immunol 2013; 34:234-42. [PMID: 23428395 DOI: 10.1016/j.it.2013.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 12/29/2012] [Accepted: 01/02/2013] [Indexed: 01/24/2023]
Abstract
The immunological synapse (IS) formed between immune cells and antigen-presenting cells (APCs) provides a platform for signaling. Protein kinase C (PKC)θ localizes in the T cell IS within the central supramolecular activation cluster (cSMAC), where it associates with CD28 and mediates T cell receptor (TCR)/CD28 signals leading to effector T (Teff) cell activation. In regulatory T (Treg) cells, PKCθ is sequestered away from the IS, and inhibits suppressive function. Other PKCs localizing in the IS mediate additional functions in various immune cells. Further work is needed to identify mechanisms underlying PKC recruitment or exclusion at the IS, potential redundancy among IS-localized PKCs, and the relevance of PKC localization for IS dynamics and lymphocyte activation.
Collapse
Affiliation(s)
- Kok-Fai Kong
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | |
Collapse
|
27
|
Michalczyk I, Sikorski AF, Kotula L, Junghans RP, Dubielecka PM. The emerging role of protein kinase Cθ in cytoskeletal signaling. J Leukoc Biol 2012. [PMID: 23192428 DOI: 10.1189/jlb.0812371] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cytoskeletal rearrangements often occur as the result of transduction of signals from the extracellular environment. Efficient awakening of this powerful machinery requires multiple activation and deactivation steps, which usually involve phosphorylation or dephosphorylation of different signaling units by kinases and phosphatases, respectively. In this review, we discuss the signaling characteristics of one of the nPKC isoforms, PKCθ, focusing on PKCθ-mediated signal transduction to cytoskeletal elements, which results in cellular rearrangements critical for cell type-specific responses to stimuli. PKCθ is the major PKC isoform present in hematopoietic and skeletal muscle cells. PKCθ plays roles in T cell signaling through the IS, survival responses in adult T cells, and T cell FasL-mediated apoptosis, all of which involve cytoskeletal rearrangements and relocation of this enzyme. PKCθ has been linked to the regulation of cell migration, lymphoid cell motility, and insulin signaling and resistance in skeletal muscle cells. Additional roles were suggested for PKCθ in mitosis and cell-cycle regulation. Comprehensive understanding of cytoskeletal regulation and the cellular "modus operandi" of PKCθ holds promise for improving current therapeutic applications aimed at autoimmune diseases.
Collapse
Affiliation(s)
- Izabela Michalczyk
- Laboratory of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | | | | | | | | |
Collapse
|
28
|
Guzman A, Zelman-Femiak M, Boergermann JH, Paschkowsky S, Kreuzaler PA, Fratzl P, Harms GS, Knaus P. SMAD versus non-SMAD signaling is determined by lateral mobility of bone morphogenetic protein (BMP) receptors. J Biol Chem 2012; 287:39492-504. [PMID: 22961979 DOI: 10.1074/jbc.m112.387639] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Bone (or body) morphogenetic proteins (BMPs) belong to the TGFβ superfamily and are crucial for embryonic patterning and organogenesis as well as for adult tissue homeostasis and repair. Activation of BMP receptors by their ligands leads to induction of several signaling cascades. Using fluorescence recovery after photobleaching, FRET, and single particle tracking microscopy, we demonstrate that BMP receptor type I and II (BMPRI and BMPRII) have distinct lateral mobility properties within the plasma membrane, which is mandatory for their involvement in different signaling pathways. Before ligand binding, BMPRI and a subpopulation of BMPRII exhibit confined motion, reflecting preassembled heteromeric receptor complexes. A second free diffusing BMPRII population only becomes restricted after ligand addition. This paper visualizes time-resolved BMP receptor complex formation and demonstrates that the lateral mobility of BMPRI has a major impact in stabilizing heteromeric BMPRI-BMPRII receptor complexes to differentially stimulate SMAD versus non-SMAD signaling.
Collapse
Affiliation(s)
- Asja Guzman
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Thielallee 63, 14195 Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Ksionda O, Saveliev A, Köchl R, Rapley J, Faroudi M, Smith-Garvin JE, Wülfing C, Rittinger K, Carter T, Tybulewicz VLJ. Mechanism and function of Vav1 localisation in TCR signalling. J Cell Sci 2012; 125:5302-14. [PMID: 22956543 PMCID: PMC3561853 DOI: 10.1242/jcs.105148] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The antigen-specific binding of T cells to antigen presenting cells results in recruitment of signalling proteins to microclusters at the cell-cell interface known as the immunological synapse (IS). The Vav1 guanine nucleotide exchange factor plays a critical role in T cell antigen receptor (TCR) signalling, leading to the activation of multiple pathways. We now show that it is recruited to microclusters and to the IS in primary CD4+ and CD8+ T cells. Furthermore, we show that this recruitment depends on the SH2 and C-terminal SH3 (SH3B) domains of Vav1, and on phosphotyrosines 112 and 128 of the SLP76 adaptor protein. Biophysical measurements show that Vav1 binds directly to these residues on SLP76 and that efficient binding depends on the SH2 and SH3B domains of Vav1. Finally, we show that the same two domains are critical for the phosphorylation of Vav1 and its signalling function in TCR-induced calcium flux. We propose that Vav1 is recruited to the IS by binding to SLP76 and that this interaction is critical for the transduction of signals leading to calcium flux.
Collapse
Affiliation(s)
- Olga Ksionda
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Anel A, Aguiló JI, Catalán E, Garaude J, Rathore MG, Pardo J, Villalba M. Protein Kinase C-θ (PKC-θ) in Natural Killer Cell Function and Anti-Tumor Immunity. Front Immunol 2012; 3:187. [PMID: 22783260 PMCID: PMC3389606 DOI: 10.3389/fimmu.2012.00187] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 06/15/2012] [Indexed: 12/24/2022] Open
Abstract
The protein kinase C-θ (PKCθ), which is essential for T cell function and survival, is also required for efficient anti-tumor immune surveillance. Natural killer (NK) cells, which express PKCθ, play a prominent role in this process, mainly by elimination of tumor cells with reduced or absent major histocompatibility complex class-I (MHC-I) expression. This justifies the increased interest of the use of activated NK cells in anti-tumor immunotherapy in the clinic. The in vivo development of MHC-I-deficient tumors is much favored in PKCθ−/− mice compared with wild-type mice. Recent data offer some clues on the mechanism that could explain the important role of PKCθ in NK cell-mediated anti-tumor immune surveillance: some studies show that PKCθ is implicated in signal transduction and anti-tumoral activity of NK cells elicited by interleukin (IL)-12 or IL-15, while others show that it is implicated in NK cell functional activation mediated by certain killer-activating receptors. Alternatively, the possibility that PKCθ is involved in NK cell degranulation is discussed, since recent data indicate that it is implicated in microtubule-organizing center polarization to the immune synapse in CD4+ T cells. The implication of PKC isoforms in degranulation has been more extensively studied in cytotoxic T lymphocyte, and these studies will be also summarized.
Collapse
Affiliation(s)
- Alberto Anel
- Departamento de Bioquímica y Biología Molecular y Celular, Universidad de Zaragoza Zaragoza, Spain
| | | | | | | | | | | | | |
Collapse
|
31
|
Nishikimi A, Uruno T, Duan X, Cao Q, Okamura Y, Saitoh T, Saito N, Sakaoka S, Du Y, Suenaga A, Kukimoto-Niino M, Miyano K, Gotoh K, Okabe T, Sanematsu F, Tanaka Y, Sumimoto H, Honma T, Yokoyama S, Nagano T, Kohda D, Kanai M, Fukui Y. Blockade of Inflammatory Responses by a Small-Molecule Inhibitor of the Rac Activator DOCK2. ACTA ACUST UNITED AC 2012; 19:488-97. [DOI: 10.1016/j.chembiol.2012.03.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 02/17/2012] [Accepted: 03/01/2012] [Indexed: 01/08/2023]
|
32
|
Acosta YY, Ojeda G, Zafra MP, Bernardone IS, Sánchez A, Dianzani U, Portolés P, Rojo JM. Dissociation of actin polymerization and lipid raft accumulation by ligation of the Inducible Costimulator (ICOS, CD278). ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.inmuno.2011.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
33
|
Sugiyama H, Kashimoto T, Ueno S, Ehara H, Kodama T, Iida T, Susa N. Relationship between localization on cellular membranes and cytotoxicity of Vibrio vulnificus hemolysin. PLoS One 2011; 6:e26018. [PMID: 22028805 PMCID: PMC3197612 DOI: 10.1371/journal.pone.0026018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 09/15/2011] [Indexed: 12/29/2022] Open
Abstract
Vibrio vulnificus secretes a hemolysin/cytolysin (VVH) that induces cytolysis in target cells. A detergent resistant membrane domain (DRM) fraction of the cells after sucrose gradient centrifugation includes cholesterol-rich membrane microdomains which have been called “lipid rafts”. It was reported that some pore-forming toxins require association with DRM and/or lipid rafts to exert their cytotoxicity. It has also been thought that cellular cholesterol is involved in VVH cytotoxicity because VVH cytotoxicity was inhibited by pre-incubation with cholesterol. However, both cellular localization and mode of action of VVH cytotoxicity remain unclear. In this study, we investigated the relationship between VVH localization on the cellular membrane and its cytotoxicity. Oligomers of VVH were detected from DRM fractions by sucrose gradient ultracentrifugation but all of these oligomers shifted from DRM fractions to non-DRM fractions after treatment with methyl-beta-cyclodextrin (MβCD), a cholesterol sequestering agent. On the other hand, immunofluorescence analysis showed that VVH did not co-localize with major lipid raft markers on cellular membrane of CHO cells. These data suggested that VVH localized at membrane regions which are relatively abundant in cholesterol but which are not identical with lipid rafts. To determine the linkage between localization and cytotoxicity of VVH, cytotoxicity was evaluated in MβCD-treated CHO cells. The cytotoxicity of VVH was not decreased by the MβCD treatment. In addition, the amount of VVH oligomer did not decrease in MβCD-treated CHO cells. Thus, we found that the amount of oligomer on cellular membrane is important for induction of cytotoxicity, whereas localization to lipid rafts on the cellular membrane was not essential to cytotoxicity.
Collapse
Affiliation(s)
- Hiroyuki Sugiyama
- Laboratory of Veterinary Public Health, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Takashige Kashimoto
- Laboratory of Veterinary Public Health, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
- * E-mail:
| | - Shunji Ueno
- Laboratory of Veterinary Public Health, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Hayato Ehara
- Laboratory of Veterinary Public Health, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Toshio Kodama
- Department of Bacterial Infections, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Tetsuya Iida
- International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Nobuyuki Susa
- Laboratory of Veterinary Public Health, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| |
Collapse
|
34
|
Gibson AW, Li X, Wu J, Baskin JG, Raman C, Edberg JC, Kimberly RP. Serine phosphorylation of FcγRI cytoplasmic domain directs lipid raft localization and interaction with protein 4.1G. J Leukoc Biol 2011; 91:97-103. [PMID: 22003208 DOI: 10.1189/jlb.0711368] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The high-affinity IgG receptor (CD64, FcγRI) has several special capacities, including the receptor-stimulated cleavage of the cell surface B cell-activating factor of the TNF superfamily (TNFSF13B). With the use of the yeast two-hybrid system, we and others have shown that FcγRI interacts with protein 4.1G (EPB41L2). Our mutational analyses identified two required 4.1G-interacting regions in the FcγRI CY and one FcγRI-interacting site in the C-terminus of protein 4.1G. Herein, we explore mechanism(s) that may regulate the interaction between protein 4.1G and FcγRI CY and influence FcγRI membrane mobility and function. We show that FcγRI CY interacts with protein 4.1G in vitro and that FcγRI coimmunoprecipitates protein 4.1G in freshly isolated human PBMC. With the use of immunostaining, we show that FcγRI colocalizes with protein 4.1G in unstimulated U937 cells, in which the FcγRI CY is constitutively serine-phosphorylated, but significant uncoupling occurs following FcγRI cross-linking, suggesting phosphoserine-regulated interaction. In vitro, protein 4.1G interacted preferentially with CK2-phosphorylated FcγRI CY, and compared with WT FcγRI, a nonphosphorylatable FcγRI mutant receptor was excluded from lipid rafts, suggesting a key role for protein 4.1G in targeting phosphorylated FcγRI to rafts. These data are consistent with a phosphoserine-dependent tethering role for protein 4.1G in maintaining FcγRI in lipid rafts and provide insight into the unique phosphoserine-based regulation of receptor signaling by FcγRI CY.
Collapse
Affiliation(s)
- Andrew W Gibson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Cornely R, Rentero C, Enrich C, Grewal T, Gaus K. Annexin A6 is an organizer of membrane microdomains to regulate receptor localization and signalling. IUBMB Life 2011; 63:1009-17. [PMID: 21990038 DOI: 10.1002/iub.540] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Accepted: 06/16/2011] [Indexed: 12/13/2022]
Abstract
Annexin A6 (AnxA6) belongs to the conserved annexin protein family--a group of Ca(2+) -dependent membrane binding proteins. It is the largest of all annexin proteins and upon activation, binds to negatively charged phospholipids in the plasma membrane and endosomes. In addition, AnxA6 associates with cholesterol-rich membrane microdomains termed lipid rafts. Membrane cholesterol triggers Ca(2+) -independent translocation of AnxA6 to membranes and AnxA6 levels determine the number of caveolae, a form of specialized rafts at the cell surface. AnxA6 also has an F-actin binding domain and interacts with cytoskeleton components. Taken together, this suggests that AnxA6 has a scaffold function to link membrane microdomains with the organization of the cytoskeleton. Such a link facilitates AnxA6 to participate in plasma membrane repair and it would also impact on receptor signalling at the cell surface, growth factor, and lipoprotein receptor trafficking, Ca(2+) -channel activity and T cell activation. Hence, the regulation of cell surface receptors by AnxA6 may be facilitated by its unique structure that allows recruitment of interaction partners and simultaneously bridging specialized membrane domains with cortical actin surrounding activated receptors.
Collapse
Affiliation(s)
- Rhea Cornely
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | | | | | | | | |
Collapse
|
36
|
Rahaman SO, Zhou G, Silverstein RL. Vav protein guanine nucleotide exchange factor regulates CD36 protein-mediated macrophage foam cell formation via calcium and dynamin-dependent processes. J Biol Chem 2011; 286:36011-36019. [PMID: 21865158 DOI: 10.1074/jbc.m111.265082] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Atherosclerosis, a chronic inflammatory disease, results in part from the accumulation of modified lipoproteins in the arterial wall and formation of lipid-laden macrophages, known as "foam cells." Recently, we reported that CD36, a scavenger receptor, contributes to activation of Vav-family guanine nucleotide exchange factors by oxidatively modified LDL in macrophages. We also discovered that CD36-dependent uptake of oxidized LDL (oxLDL) in vitro and foam cell formation in vitro and in vivo was significantly reduced in macrophages deficient of Vav proteins. The goal of the present study was to identify the mechanisms by which Vav proteins regulate CD36-dependent foam cell formation. We now show that a Vav-dynamin signaling axis plays a critical role in generating calcium signals in mouse macrophages exposed to CD36-specific oxidized phospholipid ligands. Chelation of intracellular Ca(2+) or inhibition of phospholipase C-γ (PLC-γ) inhibited Vav activation (85 and 70%, respectively, compared with vehicle control) and reduced foam cell formation (approximately 75%). Knockdown of expression by siRNA or inhibition of GTPase activity of dynamin 2, a Vav-interacting protein involved in endocytic vesicle fission, significantly blocked oxLDL uptake and inhibited foam cell formation. Immunofluorescence microscopy studies showed that Vav1 and dynamin 2 colocalized with internalized oxLDL in macrophages and that activation and mobilization of dynamin 2 by oxLDL was impaired in vav null cells. These studies identified previously unknown components of the CD36 signaling pathway, demonstrating that Vav proteins regulate oxLDL uptake and foam cell formation via calcium- and dynamin 2-dependent processes and thus represent novel therapeutic targets for atherosclerosis.
Collapse
Affiliation(s)
- S Ohidar Rahaman
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195.
| | - Gang Zhou
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Roy L Silverstein
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio 44195.
| |
Collapse
|
37
|
Choi S, Schwartz RH. Impairment of immunological synapse formation in adaptively tolerant T cells. THE JOURNAL OF IMMUNOLOGY 2011; 187:805-16. [PMID: 21685322 DOI: 10.4049/jimmunol.1003314] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adaptive tolerance is a hyporesponsive state in which lymphocyte Ag receptor signaling becomes desensitized after prolonged in vivo encounter with Ag. The molecular mechanisms underlying this hyporesponsive state in T cells are not fully understood, although a major signaling block has been shown to be present at the level of ZAP70 phosphorylation of linker for activation of T cells (LAT). In this study, we investigated the ability of adaptively tolerant mouse T cells to form conjugates with Ag-bearing APCs and to translocate signaling molecules into the interface between the T cells and APCs. Compared with naive or preactivated T cells, adaptively tolerant T cells showed no dramatic impairment in their formation of conjugates with APCs. In contrast, there was a large impairment in immunological synapse formation. Adaptively tolerant T cells were defective in their translocation of signaling molecules, such as ZAP70, LAT, and phospholipase C γ1, into the T cell-APC contact sites. Although Ag-induced activation of VAV1 was normal, VAV's recruitment into the synapse was also impaired. Interestingly, expressions of both IL-2-inducible T cell kinase and growth factor receptor-bound protein 2-related adaptor downstream of SHC were decreased by 60-80% in adaptively tolerant T cells. These decreases, in addition to the impairment in LAT phosphorylation by ZAP70, appear to be the major impediments to the phosphorylation of SLP76 (SRC homology 2 domain-containing leukocyte protein of 76 kDa) and the recruitment of VAV1, which are important for stable immunological synapse formation.
Collapse
Affiliation(s)
- Seeyoung Choi
- Laboratory of Cellular and Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-0420, USA
| | | |
Collapse
|
38
|
T-cell receptor ligation induces distinct signaling pathways in naive vs. antigen-experienced T cells. Proc Natl Acad Sci U S A 2011; 108:1549-54. [PMID: 21205892 DOI: 10.1073/pnas.1017340108] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Naïve T lymphocytes display weaker and slower responses than antigen-experienced cells for reasons that are not well understood. Here we show that T-cell receptor (TCR) stimulation induces distinct ERK and p38 phosphorylation patterns in naïve and antigen-experienced human T cells, and that these contribute to the differential responses shown by these cells. Specifically, TCR ligation triggers the activation of the ERK pathway in naïve cells. This phosphorylation of ERK attenuates subsequent calcium influx and accelerates the degradation of the signalsome. In contrast, anti-CD3 stimulation of experienced cells results in the phosphorylation of p38 via an association with Discs large (Dlg). Thus, there are distinct signaling pathways triggered by TCR ligation that impair signaling in naïve cells and facilitate it in antigen-experienced cells.
Collapse
|
39
|
Chae HD, Siefring JE, Hildeman DA, Gu Y, Williams DA. RhoH regulates subcellular localization of ZAP-70 and Lck in T cell receptor signaling. PLoS One 2010; 5:e13970. [PMID: 21103055 PMCID: PMC2980477 DOI: 10.1371/journal.pone.0013970] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Accepted: 10/19/2010] [Indexed: 01/26/2023] Open
Abstract
RhoH is an hematopoietic-specific, GTPase-deficient Rho GTPase that plays a role in T development. We investigated the mechanisms of RhoH function in TCR signaling. We found that the association between Lck and CD3ζ was impaired in RhoH-deficient T cells, due to defective translocation of both Lck and ZAP-70 to the immunological synapse. RhoH with Lck and ZAP-70 localizes in the detergent-soluble membrane fraction where the complex is associated with CD3ζ phosphorylation. To determine if impaired translocation of ZAP-70 was a major determinant of defective T cell development, Rhoh(-/-) bone marrow cells were transduced with a chimeric myristoylation-tagged ZAP-70. Myr-ZAP-70 transduced cells partially reversed the in vivo defects of RhoH-associated thymic development and TCR signaling. Together, our results suggest that RhoH regulates TCR signaling via recruitment of ZAP-70 and Lck to CD3ζ in the immunological synapse. Thus, we define a new function for a RhoH GTPase as an adaptor molecule in TCR signaling pathway.
Collapse
Affiliation(s)
- Hee-Don Chae
- Division of Experimental Hematology, Cincinnati Children's Research Foundation, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Jamie E. Siefring
- Division of Experimental Hematology, Cincinnati Children's Research Foundation, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - David A. Hildeman
- Division of Immunobiology, Cincinnati Children's Research Foundation, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Yi Gu
- Division of Experimental Hematology, Cincinnati Children's Research Foundation, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - David A. Williams
- Division of Experimental Hematology, Cincinnati Children's Research Foundation, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
40
|
Guanine nucleotide exchange factors for RhoGTPases: good therapeutic targets for cancer therapy? Cell Signal 2010; 23:969-79. [PMID: 21044680 DOI: 10.1016/j.cellsig.2010.10.022] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 10/23/2010] [Indexed: 12/12/2022]
Abstract
Rho guanosine triphosphatases (GTPases) are a family of small proteins which function as molecular switches in a variety of signaling pathways following stimulation of cell surface receptors. RhoGTPases regulate numerous cellular processes including cytoskeleton organization, gene transcription, cell proliferation, migration, growth and cell survival. Because of their central role in regulating processes that are dysregulated in cancer, it seems reasonable that defects in the RhoGTPase pathway may be involved in the development of cancer. RhoGTPase activity is regulated by a number of protein families: guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs) and guanine nucleotide-dissociation inhibitors (GDIs). This review discusses the participation of RhoGTPases and their regulators, especially GEFs in human cancers. In particular, we focus on the involvement of the RhoGTPase GEF, Vav1, a hematopoietic specific signal transducer which is involved in human neuroblastoma, pancreatic ductal carcinoma and lung cancer. Finally, we summarize recent advances in the design and application of a number of molecules that specifically target individual RhoGTPases or their regulators or effectors, and discuss their potential for cancer therapy.
Collapse
|
41
|
The plasma membrane plays a central role in cells response to mechanical stress. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2010; 1798:1739-49. [DOI: 10.1016/j.bbamem.2010.06.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 06/07/2010] [Accepted: 06/09/2010] [Indexed: 11/20/2022]
|
42
|
Radhakrishnan A, Yeo D, Brown G, Myaing MZ, Iyer LR, Fleck R, Tan BH, Aitken J, Sanmun D, Tang K, Yarwood A, Brink J, Sugrue RJ. Protein analysis of purified respiratory syncytial virus particles reveals an important role for heat shock protein 90 in virus particle assembly. Mol Cell Proteomics 2010; 9:1829-48. [PMID: 20530633 DOI: 10.1074/mcp.m110.001651] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In this study, we used imaging and proteomics to identify the presence of virus-associated cellular proteins that may play a role in respiratory syncytial virus (RSV) maturation. Fluorescence microscopy of virus-infected cells revealed the presence of virus-induced cytoplasmic inclusion bodies and mature virus particles, the latter appearing as virus filaments. In situ electron tomography suggested that the virus filaments were complex structures that were able to package multiple copies of the virus genome. The virus particles were purified, and the protein content was analyzed by one-dimensional nano-LC MS/MS. In addition to all the major virus structural proteins, 25 cellular proteins were also detected, including proteins associated with the cortical actin network, energy pathways, and heat shock proteins (HSP70, HSC70, and HSP90). Representative actin-associated proteins, HSC70, and HSP90 were selected for further biological validation. The presence of beta-actin, filamin-1, cofilin-1, HSC70, and HSP90 in the virus preparation was confirmed by immunoblotting using relevant antibodies. Immunofluorescence microscopy of infected cells stained with antibodies against relevant virus and cellular proteins confirmed the presence of these cellular proteins in the virus filaments and inclusion bodies. The relevance of HSP90 to virus infection was examined using the specific inhibitors 17-N-Allylamino-17-demethoxygeldanamycin. Although virus protein expression was largely unaffected by these drugs, we noted that the formation of virus particles was inhibited, and virus transmission was impaired, suggesting an important role for HSP90 in virus maturation. This study highlights the utility of proteomics in facilitating both our understanding of the role that cellular proteins play during RSV maturation and, by extrapolation, the identification of new potential targets for antiviral therapy.
Collapse
Affiliation(s)
- Anuradha Radhakrishnan
- Division of Molecular and Cell Biology, School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Luo J, Xu T, Wang X, Ba X, Feng X, Deepak V, Zeng X. PI3K is involved in L-selectin- and PSGL-1-mediated neutrophil rolling on E-selectin via F-actin redistribution and assembly. J Cell Biochem 2010; 110:910-9. [DOI: 10.1002/jcb.22603] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
44
|
Chichili GR, Westmuckett AD, Rodgers W. T cell signal regulation by the actin cytoskeleton. J Biol Chem 2010; 285:14737-46. [PMID: 20194498 DOI: 10.1074/jbc.m109.097311] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
T cells form an immunological synapse (IS) that sustains and regulates signals for cell stimulation. Enriched in the IS is the Src family kinase Lck. Conversely, the membrane phosphatase CD45, which activates Src family kinases, is excluded, and this is necessary to avoid quenching of T cell receptor phosphosignals. Data suggest that this arrangement occurs by an enrichment of cholesterol-dependent rafts in the IS. However, the role of rafts in structuring the IS remains unclear. To address this question, we used fluorescence resonance energy transfer (FRET) to interrogate the nanoscopic structure of the IS. The FRET probes consisted of membrane-anchored fluorescent proteins with distinct affinities for rafts. Both the raft and nonraft probes exhibited clustering in the IS. However, co-clustering of raft donor-acceptor pairs was 10-fold greater than co-clustering of raft-nonraft pairs. We measured the effect of disrupting rafts in the IS on CD45 localization and Lck regulation by treating stimulated T cells with filipin. The filipin specifically disrupted co-clustering of the raft FRET pairs in the IS and allowed targeting of CD45 to the IS and dephosphorylation of the regulatory tyrosine of Lck. Clustering of the raft probes was also sensitive to latrunctulin B, which disrupts actin filaments. Strikingly, enriching the cortical cytoskeleton using jasplakinolide maintained raft probe co-clustering, CD45 exclusion, and Lck regulation in the IS following the addition of filipin. These data show the actin cytoskeleton maintains a membrane raft environment in the IS that promotes Lck regulation by excluding CD45.
Collapse
Affiliation(s)
- Gurunadh R Chichili
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | | | | |
Collapse
|
45
|
Vav1 couples the T cell receptor to cAMP response element activation via a PKC-dependent pathway. Cell Signal 2010; 22:944-54. [PMID: 20138987 DOI: 10.1016/j.cellsig.2010.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 01/25/2010] [Accepted: 02/01/2010] [Indexed: 12/21/2022]
Abstract
The transcription factor cAMP-responsive element binding protein (CREB) is a regulator of the expression of several genes important for lymphocyte activation and proliferation. However, the proximal signaling events leading to activation of CREB in T cells upon antigen receptor stimulation remain unknown. Here we identify a role for Vav1 in the activation of the cAMP response element (CRE), the binding site for CREB. T cell receptor (TCR)/CD28 - induced costimulation of Jurkat T cells expressing Vav1 but not a GEF-deficient mutant showed increased CRE activation (7.2+/-2.4 fold over control), whereas Vav1 downregulation by siRNA reduced activation of CRE by 2.6+/-1.3 fold. Inhibition of PKC and MEK but not p38 could reduce Vav1-mediated CRE activation, suggesting that Vav1 transmits TCR and CD28 signals to activation of CRE via PKC and ERK signaling pathways. As a consequence, downregulation of Vav1 impaired the expression of several CRE-containing genes like cyclin D1, INFgamma and IL-2, whereas overexpression of Vav1 enhanced CRE-dependent gene expression. Furthermore, cAMP-induced CRE-dependent transcription and gene expression was also modulated by Vav1, but did not require activation of PKC and the GEF function of Vav1. Our data provide insights into the signal transduction events regulating CRE-mediated gene expression in T cells, which affects T cell development, proliferation and activation. We identify Vav1 as an essential component of TCR-induced CRE activation and gene expression, which underlines the central role for Vav1 as key player for TCR signal transduction and gene expression.
Collapse
|
46
|
Munday AD, Gaus K, López JA. The platelet glycoprotein Ib-IX-V complex anchors lipid rafts to the membrane skeleton: implications for activation-dependent cytoskeletal translocation of signaling molecules. J Thromb Haemost 2010; 8:163-72. [PMID: 19874464 DOI: 10.1111/j.1538-7836.2009.03656.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND The glycoprotein (GP) Ib-IX-V complex attaches platelets to areas of endothelial damage by binding von Willebrand factor (VWF), an interaction that transmits intracellular activation signals. These signals require that the complex associates with both lipid rafts and the membrane cytoskeleton, but it is not clear whether the same GPIb-IX-V subpopulation associates with both structures. OBJECTIVES To determine which subpopulation of GPIb-IX-V associates with lipid rafts, and the consequences of that interaction. METHODS We analyzed the content of proteins (particularly the GPIb-IX-V complex) and lipids in rafts from detergent lysates of platelets before and after removal of the actin cytoskeleton alone or both the actin cytoskeleton and membrane skeleton (by successive centrifugations of 15,800 x g and 100,000 x g). RESULTS In unstimulated platelets, little raft-associated GPIb-IX-V sedimented with the actin skeleton; most was removed by sedimentation of the membrane skeleton. The Src family kinase Lyn followed the same pattern. In VWF-activated platelets, almost all of the GPIb-IX-V complex and Lyn in rafts sedimented with the actin cytoskeleton, consistent with a previously described crosslinking of the membrane and actin skeletal structures following platelet activation. Disruption of the GPIbalpha-filamin linkage with N-ethylmaleimide prevented depletion of raft-associated GPIb-IX-V by skeletal sedimentation. Not all raft-associated proteins and lipids followed this pattern. CONCLUSION These results suggest that the raft association and cytoskeletal linkage of the GPIb-IX-V complex are interrelated, and both are required for optimal receptor function, perhaps because raft association attracts signaling proteins and membrane skeletal association allows these proteins to move en masse to new locations.
Collapse
Affiliation(s)
- A D Munday
- Puget Sound Blood Center, and Hematology Division (Medicine), University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
47
|
Stuermer CA. The reggie/flotillin connection to growth. Trends Cell Biol 2010; 20:6-13. [DOI: 10.1016/j.tcb.2009.10.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 10/09/2009] [Accepted: 10/09/2009] [Indexed: 10/20/2022]
|
48
|
Sadegh-Nasseri S, Dalai SK, Korb Ferris LC, Mirshahidi S. Suboptimal engagement of the T-cell receptor by a variety of peptide-MHC ligands triggers T-cell anergy. Immunology 2009; 129:1-7. [PMID: 20002785 DOI: 10.1111/j.1365-2567.2009.03206.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
T cells recognize antigen via the T-cell receptor (TCR) and produce a spectrum of responses that range from activation to anergy or cell death. The variety of outcomes may be dictated by the strength of the signals transmitted upon cognate recognition of the TCR. The physiological outcome of TCR engagement is determined by several factors, including the avidity of the ligand for TCR, the duration of engagement, and the presence and nature of accessory molecules present on antigen-presenting cells (APCs). In this review, we discuss a model of anergy induced by presentation of low densities of peptide-major histocompatibility complex (MHC) ligand in CD4(+) T cells and compare it to anergy induced by altered peptide ligands in an effort to identify a unifying mechanism. We suggest that altered peptide ligand (APL) and low densities of agonist ligands induce anergy by engaging less than optimal numbers of TCRs. The physiological impacts of anergy in memory CD4(+) T cells are discussed.
Collapse
|
49
|
Oakley FD, Smith RL, Engelhardt JF. Lipid rafts and caveolin-1 coordinate interleukin-1beta (IL-1beta)-dependent activation of NFkappaB by controlling endocytosis of Nox2 and IL-1beta receptor 1 from the plasma membrane. J Biol Chem 2009; 284:33255-64. [PMID: 19801678 DOI: 10.1074/jbc.m109.042127] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recent evidence suggests that signaling by the proinflammatory cytokine interleukin-1beta (IL-1beta) is dependent on reactive oxygen species derived from NADPH oxidase. Redox signaling in response to IL-1beta is known to require endocytosis of its cognate receptor (IL-1R1) following ligand binding and the formation of redox-active signaling endosomes that contain Nox2 (also called redoxosomes). The consequent generation of reactive oxygen species by redoxosomes is responsible for the downstream recruitment of IL-1R1 effectors (IRAK, TRAF6, and IkappaB kinase kinases) and ultimately for activation of the transcription factor NFkappaB. Despite this knowledge of the signaling events that occur downstream of redoxosome formation, an understanding of the mechanisms that coordinate the genesis of redoxosomes following IL-1beta stimulation has been lacking. Here, we demonstrate that lipid rafts play an important role in this process. We show that Nox2 and IL-1R1 localize to plasma membrane lipid rafts in the unstimulated state and that IL-1beta signals caveolin-1-dependent endocytosis of both proteins into the redoxosome. We also show that inhibiting lipid raft-mediated endocytosis prevents NFkappaB activation. Finally, we demonstrate that Vav1, a Rac1 guanine exchange factor and activator of Nox2, is recruited to lipid rafts following IL-1beta stimulation and that it is required for NFkappaB activation. Our results fill in an important mechanistic gap in the understanding of early IL-1R1 and Nox2 signaling events that control NFkappaB activation, a redox-dependent process important in inflammation.
Collapse
Affiliation(s)
- Fredrick D Oakley
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | |
Collapse
|
50
|
Bhavsar PJ, Vigorito E, Turner M, Ridley AJ. Vav GEFs regulate macrophage morphology and adhesion-induced Rac and Rho activation. Exp Cell Res 2009; 315:3345-58. [PMID: 19715691 DOI: 10.1016/j.yexcr.2009.08.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 08/18/2009] [Accepted: 08/21/2009] [Indexed: 12/18/2022]
Abstract
The Vav family of proteins have the potential to act as both signalling adapters and GEFs for Rho GTPases. They have therefore been proposed as regulators of the cytoskeleton in various cell types. We have used macrophages from mice deficient in all three Vav isoforms to determine how their function affects cell morphology and migration. Macrophages lacking Vav proteins adopt an elongated morphology and have enhanced migratory persistence in culture. To investigate the pathways through which Vav proteins exert their effects we analysed the responses of macrophages to the chemoattractant CSF-1 and to adhesion. We found that morphological and signalling responses of macrophages to CSF-1 did not require Vav proteins. In contrast, adhesion-induced cell spreading, RhoA and Rac1 activation and cell signalling were all dependent on Vav proteins. We propose that Vav proteins affect macrophage morphology and motile behaviour by coupling adhesion receptors to Rac1 and RhoA activity and regulating adhesion signalling events such as paxillin and ERK1/2 phosphorylation by acting as adapters.
Collapse
Affiliation(s)
- Parag J Bhavsar
- King's College London, Randall Division of Cell and Molecular Biophysics, Guy's Campus, London, UK
| | | | | | | |
Collapse
|