1
|
Griffith AA, Callahan KP, King NG, Xiao Q, Su X, Salomon AR. SILAC Phosphoproteomics Reveals Unique Signaling Circuits in CAR-T Cells and the Inhibition of B Cell-Activating Phosphorylation in Target Cells. J Proteome Res 2022; 21:395-409. [PMID: 35014847 PMCID: PMC8830406 DOI: 10.1021/acs.jproteome.1c00735] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chimeric antigen receptor (CAR) is a single-pass transmembrane receptor designed to specifically target and eliminate cancers. While CARs prove highly efficacious against B cell malignancies, the intracellular signaling events which promote CAR T cell activity remain elusive. To gain further insight into both CAR T cell signaling and the potential signaling response of cells targeted by CAR, we analyzed phosphopeptides captured by two separate phosphoenrichment strategies from third generation CD19-CAR T cells cocultured with SILAC labeled Raji B cells by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Here, we report that CD19-CAR T cells upregulated several key phosphorylation events also observed in canonical T cell receptor (TCR) signaling, while Raji B cells exhibited a significant decrease in B cell receptor-signaling related phosphorylation events in response to coculture. Our data suggest that CD19-CAR stimulation activates a mixture of unique CD19-CAR-specific signaling pathways and canonical TCR signaling, while global phosphorylation in Raji B cells is reduced after association with the CD19-CAR T cells.
Collapse
Affiliation(s)
- Alijah A. Griffith
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI, 02912
| | - Kenneth P. Callahan
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI, 02912
| | - Nathan Gordo King
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI, 02912
| | - Qian Xiao
- Department of Cell Biology, Yale School of Medicine, Yale University, New Haven, CT, 06520
| | - Xiaolei Su
- Department of Cell Biology, Yale School of Medicine, Yale University, New Haven, CT, 06520
| | - Arthur R. Salomon
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI, 02912,
| |
Collapse
|
2
|
Lawton ML, Emili A. Mass Spectrometry-Based Phosphoproteomics and Systems Biology: Approaches to Study T Lymphocyte Activation and Exhaustion. J Mol Biol 2021; 433:167318. [PMID: 34687714 DOI: 10.1016/j.jmb.2021.167318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/04/2021] [Accepted: 10/15/2021] [Indexed: 11/24/2022]
Abstract
T lymphocytes respond to extracellular cues and recognize and clear foreign bodies. These functions are tightly regulated by receptor-mediated intracellular signal transduction pathways and phosphorylation cascades resulting in rewiring of transcription, cell adhesion, and metabolic pathways, which leads to changes in downstream effector functions including cytokine secretion and target-cell killing. Given that these pathways become dysregulated in chronic diseases such as cancer, auto-immunity, diabetes, and persistent infections, mapping T cell signaling dynamics in normal and pathological states is central to understanding and modulating immune system behavior. Despite recent advances, there remains much to be learned from the study of T cell signaling at a systems level. The application of global phospho-proteomic profiling technology has the potential to provide unprecedented insights into the molecular networks that govern T cell function. These include capturing the spatiotemporal dynamics of the T cell responses as an ensemble of interacting components, rather than a static view at a single point in time. In this review, we describe innovative experimental approaches to study signaling mechanisms in the TCR, co-stimulatory receptors, synthetic signaling molecules such as chimeric antigen receptors, inhibitory receptors, and T cell exhaustion. Technical advances in mass spectrometry and systems biology frameworks are emphasized as these are poised to identify currently unknown functional relationships and dependencies to create causal predictive models that expand from the traditional narrow reductionist lens of singular components in isolation.
Collapse
Affiliation(s)
- Matthew L Lawton
- Center for Network Systems Biology, Boston University, Boston, MA, USA; Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Andrew Emili
- Center for Network Systems Biology, Boston University, Boston, MA, USA; Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; Department of Biology, Boston University, Boston, MA, USA.
| |
Collapse
|
3
|
Lawrie J, Waldrop S, Morozov A, Niu W, Guo J. Engineering of a Small Protein Scaffold To Recognize Sulfotyrosine with High Specificity. ACS Chem Biol 2021; 16:1508-1517. [PMID: 34251168 DOI: 10.1021/acschembio.1c00382] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Protein tyrosine O-sulfation is an essential post-translational modification required for effective biological processes such as hemostasis, inflammatory response, and visual phototransduction. Because of its unstable nature under mass spectrometry conditions and residing on low-abundance cell surface proteins, sulfated tyrosine (sulfotyrosine) residues are difficult to detect or analyze. Enrichment of sulfotyrosine-containing proteins (sulfoproteins) from complex biological samples are typically required before analysis. In this work, we seek to engineer the phosphotyrosine binding pocket of a Src Homology 2 (SH2) domain to act as an antisulfotyrosine antibody mimic. Using tailored selection schemes, several SH2 mutants are identified with high affinity and specificity to sulfotyrosine. Further molecular docking simulations highlight potential mechanisms supporting observed characteristics of these SH2 mutants. Utilities of the evolved SH2 mutants were demonstrated by the detection and enrichment of sulfoproteins.
Collapse
Affiliation(s)
- Justin Lawrie
- Department of Chemistry, University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
| | - Sean Waldrop
- Department of Chemistry, University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
| | - Anya Morozov
- Department of Chemistry, University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
| | - Wei Niu
- Department of Chemical & Biomolecular Engineering, University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
| | - Jiantao Guo
- Department of Chemistry, University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
| |
Collapse
|
4
|
Locard‐Paulet M, Voisinne G, Froment C, Goncalves Menoita M, Ounoughene Y, Girard L, Gregoire C, Mori D, Martinez M, Luche H, Garin J, Malissen M, Burlet‐Schiltz O, Malissen B, Gonzalez de Peredo A, Roncagalli R. LymphoAtlas: a dynamic and integrated phosphoproteomic resource of TCR signaling in primary T cells reveals ITSN2 as a regulator of effector functions. Mol Syst Biol 2020; 16:e9524. [PMID: 32618424 PMCID: PMC7333348 DOI: 10.15252/msb.20209524] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/29/2022] Open
Abstract
T-cell receptor (TCR) ligation-mediated protein phosphorylation regulates the activation, cellular responses, and fates of T cells. Here, we used time-resolved high-resolution phosphoproteomics to identify, quantify, and characterize the phosphorylation dynamics of thousands of phosphorylation sites in primary T cells during the first 10 min after TCR stimulation. Bioinformatic analysis of the data revealed a coherent orchestration of biological processes underlying T-cell activation. In particular, functional modules associated with cytoskeletal remodeling, transcription, translation, and metabolic processes were mobilized within seconds after TCR engagement. Among proteins whose phosphorylation was regulated by TCR stimulation, we demonstrated, using a fast-track gene inactivation approach in primary lymphocytes, that the ITSN2 adaptor protein regulated T-cell effector functions. This resource, called LymphoAtlas, represents an integrated pipeline to further decipher the organization of the signaling network encoding T-cell activation. LymphoAtlas is accessible to the community at: https://bmm-lab.github.io/LymphoAtlas.
Collapse
Affiliation(s)
- Marie Locard‐Paulet
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
- Present address:
Novo Nordisk Foundation Center for Protein ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Guillaume Voisinne
- Centre d'Immunologie de Marseille‐LuminyINSERM, CNRSAix Marseille UniversitéMarseilleFrance
| | - Carine Froment
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
| | | | - Youcef Ounoughene
- Centre d'Immunologie de Marseille‐LuminyINSERM, CNRSAix Marseille UniversitéMarseilleFrance
- Centre d'ImmunophénomiqueINSERM, CNRS UMRAix Marseille UniversitéMarseilleFrance
| | - Laura Girard
- Centre d'Immunologie de Marseille‐LuminyINSERM, CNRSAix Marseille UniversitéMarseilleFrance
- Centre d'ImmunophénomiqueINSERM, CNRS UMRAix Marseille UniversitéMarseilleFrance
| | - Claude Gregoire
- Centre d'Immunologie de Marseille‐LuminyINSERM, CNRSAix Marseille UniversitéMarseilleFrance
| | - Daiki Mori
- Centre d'Immunologie de Marseille‐LuminyINSERM, CNRSAix Marseille UniversitéMarseilleFrance
- Centre d'ImmunophénomiqueINSERM, CNRS UMRAix Marseille UniversitéMarseilleFrance
| | - Manuel Martinez
- Centre d'ImmunophénomiqueINSERM, CNRS UMRAix Marseille UniversitéMarseilleFrance
| | - Hervé Luche
- Centre d'ImmunophénomiqueINSERM, CNRS UMRAix Marseille UniversitéMarseilleFrance
| | - Jerôme Garin
- CEA, BIG, Biologie à Grande Echelle, INSERM, U1038Université Grenoble‐AlpesGrenobleFrance
| | - Marie Malissen
- Centre d'Immunologie de Marseille‐LuminyINSERM, CNRSAix Marseille UniversitéMarseilleFrance
- Centre d'ImmunophénomiqueINSERM, CNRS UMRAix Marseille UniversitéMarseilleFrance
| | - Odile Burlet‐Schiltz
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
| | - Bernard Malissen
- Centre d'Immunologie de Marseille‐LuminyINSERM, CNRSAix Marseille UniversitéMarseilleFrance
- Centre d'ImmunophénomiqueINSERM, CNRS UMRAix Marseille UniversitéMarseilleFrance
| | - Anne Gonzalez de Peredo
- Institut de Pharmacologie et de Biologie Structurale (IPBS)Université de Toulouse, CNRS, UPSToulouseFrance
| | - Romain Roncagalli
- Centre d'Immunologie de Marseille‐LuminyINSERM, CNRSAix Marseille UniversitéMarseilleFrance
| |
Collapse
|
5
|
Lombard-Banek C, Schiel JE. Mass Spectrometry Advances and Perspectives for the Characterization of Emerging Adoptive Cell Therapies. Molecules 2020; 25:E1396. [PMID: 32204371 PMCID: PMC7144572 DOI: 10.3390/molecules25061396] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022] Open
Abstract
Adoptive cell therapy is an emerging anti-cancer modality, whereby the patient's own immune cells are engineered to express T-cell receptor (TCR) or chimeric antigen receptor (CAR). CAR-T cell therapies have advanced the furthest, with recent approvals of two treatments by the Food and Drug Administration of Kymriah (trisagenlecleucel) and Yescarta (axicabtagene ciloleucel). Recent developments in proteomic analysis by mass spectrometry (MS) make this technology uniquely suited to enable the comprehensive identification and quantification of the relevant biochemical architecture of CAR-T cell therapies and fulfill current unmet needs for CAR-T product knowledge. These advances include improved sample preparation methods, enhanced separation technologies, and extension of MS-based proteomic to single cells. Innovative technologies such as proteomic analysis of raw material quality attributes (MQA) and final product quality attributes (PQA) may provide insights that could ultimately fuel development strategies and lead to broad implementation.
Collapse
Affiliation(s)
- Camille Lombard-Banek
- National Institute of Standards and Technology, Gaithersburg, MD 20899, USA;
- Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
| | - John E. Schiel
- National Institute of Standards and Technology, Gaithersburg, MD 20899, USA;
- Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
| |
Collapse
|
6
|
Valdés A, Bergström Lind S. Mass Spectrometry-Based Analysis of Time-Resolved Proteome Quantification. Proteomics 2019; 20:e1800425. [PMID: 31652013 DOI: 10.1002/pmic.201800425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/20/2019] [Indexed: 11/09/2022]
Abstract
The aspect of time is essential in biological processes and thus it is important to be able to monitor signaling molecules through time. Proteins are key players in cellular signaling and they respond to many stimuli and change their expression in many time-dependent processes. Mass spectrometry (MS) is an important tool for studying proteins, including their posttranslational modifications and their interaction partners-both in qualitative and quantitative ways. In order to distinguish the different trends over time, proteins, modification sites, and interacting proteins must be compared between different time points, and therefore relative quantification is preferred. In this review, the progress and challenges for MS-based analysis of time-resolved proteome dynamics are discussed. Further, aspects on model systems, technologies, sampling frequencies, and presentation of the dynamic data are discussed.
Collapse
Affiliation(s)
- Alberto Valdés
- Department of Analytical Chemistry, Physical Chemistry and Chemical Engineering, University of Alcalá, Ctra. Madrid-Barcelona, Km. 33.600, 28871, Alcalá de Henares, Madrid, Spain
| | - Sara Bergström Lind
- Department of Chemistry-BMC, Analytical Chemistry, Uppsala University, Box 599, 75124, Uppsala, Sweden
| |
Collapse
|
7
|
Salter AI, Ivey RG, Kennedy JJ, Voillet V, Rajan A, Alderman EJ, Voytovich UJ, Lin C, Sommermeyer D, Liu L, Whiteaker JR, Gottardo R, Paulovich AG, Riddell SR. Phosphoproteomic analysis of chimeric antigen receptor signaling reveals kinetic and quantitative differences that affect cell function. Sci Signal 2018; 11:11/544/eaat6753. [PMID: 30131370 DOI: 10.1126/scisignal.aat6753] [Citation(s) in RCA: 343] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chimeric antigen receptors (CARs) link an antigen recognition domain to intracellular signaling domains to redirect T cell specificity and function. T cells expressing CARs with CD28/CD3ζ or 4-1BB/CD3ζ signaling domains are effective at treating refractory B cell malignancies but exhibit differences in effector function, clinical efficacy, and toxicity that are assumed to result from the activation of divergent signaling cascades. We analyzed stimulation-induced phosphorylation events in primary human CD8+ CD28/CD3ζ and 4-1BB/CD3ζ CAR T cells by mass spectrometry and found that both CAR constructs activated similar signaling intermediates. Stimulation of CD28/CD3ζ CARs activated faster and larger-magnitude changes in protein phosphorylation, which correlated with an effector T cell-like phenotype and function. In contrast, 4-1BB/CD3ζ CAR T cells preferentially expressed T cell memory-associated genes and exhibited sustained antitumor activity against established tumors in vivo. Mutagenesis of the CAR CD28 signaling domain demonstrated that the increased CD28/CD3ζ CAR signal intensity was partly related to constitutive association of Lck with this domain in CAR complexes. Our data show that CAR signaling pathways cannot be predicted solely by the domains used to construct the receptor and that signal strength is a key determinant of T cell fate. Thus, tailoring CAR design based on signal strength may lead to improved clinical efficacy and reduced toxicity.
Collapse
Affiliation(s)
- Alexander I Salter
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Richard G Ivey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jacob J Kennedy
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Valentin Voillet
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Anusha Rajan
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Eva J Alderman
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Uliana J Voytovich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Chenwei Lin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Daniel Sommermeyer
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Lingfeng Liu
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jeffrey R Whiteaker
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Amanda G Paulovich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Stanley R Riddell
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA. .,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
8
|
Adebayo Michael AO, Ahsan N, Zabala V, Francois-Vaughan H, Post S, Brilliant KE, Salomon AR, Sanders JA, Gruppuso PA. Proteomic analysis of laser capture microdissected focal lesions in a rat model of progenitor marker-positive hepatocellular carcinoma. Oncotarget 2018; 8:26041-26056. [PMID: 28199961 PMCID: PMC5432236 DOI: 10.18632/oncotarget.15219] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/27/2017] [Indexed: 12/19/2022] Open
Abstract
We have shown previously that rapamycin, the canonical inhibitor of the mechanistic target of rapamycin (mTOR) complex 1, markedly inhibits the growth of focal lesions in the resistant hepatocyte (Solt-Farber) model of hepatocellular carcinoma (HCC) in the rat. In the present study, we characterized the proteome of persistent, pre-neoplastic focal lesions in this model. One group was administered rapamycin by subcutaneous pellet for 3 weeks following partial hepatectomy and euthanized 4 weeks after the cessation of rapamycin. A second group received placebo pellets. Results were compared to unmanipulated control animals and to animals that underwent an incomplete Solt-Farber protocol to activate hepatic progenitor cells. Regions of formalin-fixed, paraffin-embedded tissue were obtained by laser capture microdissection (LCM). Proteomic analysis yielded 11,070 unique peptides representing 2,227 proteins. Quantitation of the peptides showed increased abundance of known HCC markers (e.g., glutathione S-transferase-P, epoxide hydrolase, 6 others) and potential markers (e.g., aflatoxin aldehyde reductase, glucose 6-phosphate dehydrogenase, 10 others) in foci from placebo-treated and rapamycin-treated rats. Peptides derived from cytochrome P450 enzymes were generally reduced. Comparisons of the rapamycin samples to normal liver and to the progenitor cell model indicated that rapamycin attenuated a loss of differentiation relative to placebo. We conclude that early administration of rapamycin in the Solt-Farber model not only inhibits the growth of pre-neoplastic foci but also attenuates the loss of differentiated function. In addition, we have demonstrated that the combination of LCM and mass spectrometry-based proteomics is an effective approach to characterize focal liver lesions.
Collapse
Affiliation(s)
- Adeola O Adebayo Michael
- Department of Pediatrics, Rhode Island Hospital and Brown University, Providence, RI, USA.,Current address: Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nagib Ahsan
- Division of Biology and Medicine, Brown University, Providence, RI, USA.,Center for Cancer Research Development, Proteomics Core Facility, Rhode Island Hospital, Providence, RI, USA
| | - Valerie Zabala
- Department of Pediatrics, Rhode Island Hospital and Brown University, Providence, RI, USA
| | | | - Stephanie Post
- Department of Environmental and Evolutionary Biology, Brown University, Providence, RI, USA
| | - Kate E Brilliant
- Center for Cancer Research Development, Proteomics Core Facility, Rhode Island Hospital, Providence, RI, USA
| | - Arthur R Salomon
- Center for Cancer Research Development, Proteomics Core Facility, Rhode Island Hospital, Providence, RI, USA.,Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Jennifer A Sanders
- Department of Pediatrics, Rhode Island Hospital and Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Philip A Gruppuso
- Department of Pediatrics, Rhode Island Hospital and Brown University, Providence, RI, USA.,Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| |
Collapse
|
9
|
Kruglova NA, Meshkova TD, Kopylov AT, Mazurov DV, Filatov AV. Constitutive and activation-dependent phosphorylation of lymphocyte phosphatase-associated phosphoprotein (LPAP). PLoS One 2017; 12:e0182468. [PMID: 28827793 PMCID: PMC5565103 DOI: 10.1371/journal.pone.0182468] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 07/19/2017] [Indexed: 12/15/2022] Open
Abstract
Lymphocyte phosphatase-associated phosphoprotein (LPAP) is a small transmembrane protein expressed exclusively in lymphocytes. LPAP is a component of a supramolecular complex composed of the phosphatase CD45, the co-receptor CD4, and the kinase Lck. In contrast to its immunologically important partners, the function of LPAP is unknown. We hypothesized that the biological role of LPAP may be determined by analyzing LPAP phosphorylation. In the present study, we identified LPAP phosphorylation sites by site-directed mutagenesis, phospho-specific antibodies, and protein immunoprecipitation coupled to mass spectrometry analysis. Our results confirmed previous reports that Ser-99, Ser-153, and Ser-163 are phosphorylated, as well as provided evidence for the phosphorylation of Ser-172. Using various SDS-PAGE techniques, we detected and quantified a set of LPAP phosphoforms that were assigned to a combination of particular phosphorylation events. The phosphorylation of LPAP appears to be a tightly regulated process. Our results support the model: following phorbol 12-myristate 13-acetate (PMA) or TCR/CD3 activation of T cells, LPAP is rapidly dephosphorylated at Ser-99 and Ser-172 and slowly phosphorylated at Ser-163. Ser-153 exhibited a high basal level of phosphorylation in both resting and activated cells. Therefore, we suggest that LPAP may function as a co-regulator of T-cell signaling.
Collapse
|
10
|
Álvarez-Salamero C, Castillo-González R, Navarro MN. Lighting Up T Lymphocyte Signaling with Quantitative Phosphoproteomics. Front Immunol 2017; 8:938. [PMID: 28848546 PMCID: PMC5552657 DOI: 10.3389/fimmu.2017.00938] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/21/2017] [Indexed: 12/31/2022] Open
Abstract
Phosphorylation is the most abundant post-translational modification, regulating several aspects of protein and cell function. Quantitative phosphoproteomics approaches have expanded the scope of phosphorylation analysis enabling the quantification of changes in thousands of phosphorylation sites simultaneously in two or more conditions. These approaches offer a global view of the impact of cellular perturbations such as extracellular stimuli or gene ablation in intracellular signaling networks. Such great potential also brings on a new challenge: to identify, among the thousands of phosphorylations found in global phosphoproteomics studies, the small subset of site-specific phosphorylations expected to be functionally relevant. This review focus on updating and integrating findings on T lymphocyte signaling generated using global phosphoproteomics approaches, drawing attention on the biological relevance of the obtained data.
Collapse
Affiliation(s)
- Candelas Álvarez-Salamero
- Departamento de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain
| | | | - María N Navarro
- Departamento de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain.,Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
11
|
Lopez CE, Sheehan HC, Vierra DA, Azzinaro PA, Meedel TH, Howlett NG, Irvine SQ. Proteomic responses to elevated ocean temperature in ovaries of the ascidian Ciona intestinalis. Biol Open 2017; 6:943-955. [PMID: 28500033 PMCID: PMC5550911 DOI: 10.1242/bio.024786] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 05/10/2017] [Indexed: 11/21/2022] Open
Abstract
Ciona intestinalis, a common sea squirt, exhibits lower reproductive success at the upper extreme of the water temperatures it experiences in coastal New England. In order to understand the changes in protein expression associated with elevated temperatures, and possible response to global temperature change, we reared C. intestinalis from embryos to adults at 18°C (a temperature at which they reproduce normally at our collection site in Rhode Island) and 22°C (the upper end of the local temperature range). We then dissected ovaries from animals at each temperature, extracted protein, and measured proteomic levels using shotgun mass spectrometry (LC-MS/MS). 1532 proteins were detected at a 1% false discovery rate present in both temperature groups by our LC-MS/MS method. 62 of those proteins are considered up- or down-regulated according to our statistical criteria. Principal component analysis shows a clear distinction in protein expression pattern between the control (18°C) group and high temperature (22°C) group. Similar to previous studies, cytoskeletal and chaperone proteins are upregulated in the high temperature group. Unexpectedly, we find evidence that proteolysis is downregulated at the higher temperature. We propose a working model for the high temperature response in C. intestinalis ovaries whereby increased temperature induces upregulation of signal transduction pathways involving PTPN11 and CrkL, and activating coordinated changes in the proteome especially in large lipid transport proteins, cellular stress responses, cytoskeleton, and downregulation of energy metabolism.
Collapse
Affiliation(s)
- Chelsea E Lopez
- Departments of Biological Sciences, University of Rhode Island, Kingston, RI 02881, USA
| | - Hannah C Sheehan
- Departments of Biological Sciences, University of Rhode Island, Kingston, RI 02881, USA
| | - David A Vierra
- Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA
| | - Paul A Azzinaro
- Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA
| | - Thomas H Meedel
- Biology Department, Rhode Island College, Providence, RI 02908, USA
| | - Niall G Howlett
- Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA
| | - Steven Q Irvine
- Departments of Biological Sciences, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
12
|
Belmont J, Gu T, Mudd A, Salomon AR. A PLC-γ1 Feedback Pathway Regulates Lck Substrate Phosphorylation at the T-Cell Receptor and SLP-76 Complex. J Proteome Res 2017. [PMID: 28644030 DOI: 10.1021/acs.jproteome.6b01026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Phospholipase C gamma 1 (PLC-γ1) occupies a critically important position in the T-cell signaling pathway. While its functions as a regulator of both Ca2+ signaling and PKC-family kinases are well characterized, PLC-γ1's role in the regulation of early T-cell receptor signaling events is incompletely understood. Activation of the T-cell receptor leads to the formation of a signalosome complex between SLP-76, LAT, PLC-γ1, Itk, and Vav1. Recent studies have revealed the existence of both positive and negative feedback pathways from SLP-76 to the apical kinase in the pathway, Lck. To determine if PLC-γ1 contributes to the regulation of these feedback networks, we performed a quantitative phosphoproteomic analysis of PLC-γ1-deficient T cells. These data revealed a previously unappreciated role for PLC-γ1 in the positive regulation of Zap-70 and T-cell receptor tyrosine phosphorylation. Conversely, PLC-γ1 negatively regulated the phosphorylation of SLP-76-associated proteins, including previously established Lck substrate phosphorylation sites within this complex. While the positive and negative regulatory phosphorylation sites on Lck were largely unchanged, Tyr192 phosphorylation was elevated in Jgamma1. The data supports a model wherein Lck's targeting, but not its kinase activity, is altered by PLC-γ1, possibly through Lck Tyr192 phosphorylation and increased association of the kinase with protein scaffolds SLP-76 and TSAd.
Collapse
Affiliation(s)
- Judson Belmont
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University , Providence, Rhode Island 02912, United States
| | - Tao Gu
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University , Providence, Rhode Island 02912, United States
| | - Ashley Mudd
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University , Providence, Rhode Island 02912, United States
| | - Arthur R Salomon
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University , Providence, Rhode Island 02912, United States.,Department of Chemistry, Brown University , Providence, Rhode Island 02912, United States
| |
Collapse
|
13
|
Ahsan N, Belmont J, Chen Z, Clifton JG, Salomon AR. Highly reproducible improved label-free quantitative analysis of cellular phosphoproteome by optimization of LC-MS/MS gradient and analytical column construction. J Proteomics 2017. [PMID: 28634120 DOI: 10.1016/j.jprot.2017.06.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Expanding the sequencing depth of the peptides with a statistically significant quantitative change derived from a biological stimulation is critical. Here we demonstrate that optimization of LC gradient and analytical column construction can reveal over 30,000 unique peptides and 23,000 phosphopeptides at high confidence. The quantitative reproducibility of different analytical workflows was evaluated by comparing the phosphoproteome of CD3/4 stimulated and unstimulated T-cells as a model system. A fritless, 50cm-long column packed with 1.9μm particles operated with a standard pressure HPLC significantly improved the sequencing depth 51% and decreased the selected ion chromatogram peak spreading. Most importantly, under the optimal workflow we observed an improvement of over 300% in detection of significantly changed phosphopeptides in the stimulated cells compared with the other workflows. The discovery power of the optimized column configuration was illustrated by identification of significantly altered phosphopeptides harboring novel sites from proteins previously established as important in T cell signaling including A-Raf, B-Raf, c-Myc, CARMA1, Fyn, ITK, LAT, NFAT1/2/3, PKCα, PLCγ1/2, RAF1, and SOS1. Taken together, our results reveal the analytical power of optimized chromatography using sub 2μm particles for the analysis of the T cell phosphoproteome to reveal a vast landscape of significantly altered phosphorylation changes in response to T cell receptor stimulation.
Collapse
Affiliation(s)
- Nagib Ahsan
- Division of Biology and Medicine, Brown University, Providence, RI 02903, USA; Center for Cancer Research Development, Proteomics Core Facility, Rhode Island Hospital, Providence, RI 02903, USA
| | - Judson Belmont
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Zhuo Chen
- Department of Chemistry, Brown University, Providence, RI 02912, USA
| | - James G Clifton
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown, University, Providence, RI 02912, USA
| | - Arthur R Salomon
- Center for Cancer Research Development, Proteomics Core Facility, Rhode Island Hospital, Providence, RI 02903, USA; Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA; Department of Chemistry, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
14
|
Hatano A, Matsumoto M, Nakayama KI. Phosphoproteomics analyses show subnetwork systems in T-cell receptor signaling. Genes Cells 2016; 21:1095-1112. [DOI: 10.1111/gtc.12406] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/02/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Atsushi Hatano
- Department of Molecular and Cellular Biology; Medical Institute of Bioregulation; Kyushu University; 3-1-1 Maidashi Higashi-ku Fukuoka 812-8582 Japan
| | - Masaki Matsumoto
- Department of Molecular and Cellular Biology; Medical Institute of Bioregulation; Kyushu University; 3-1-1 Maidashi Higashi-ku Fukuoka 812-8582 Japan
| | - Keiichi I. Nakayama
- Department of Molecular and Cellular Biology; Medical Institute of Bioregulation; Kyushu University; 3-1-1 Maidashi Higashi-ku Fukuoka 812-8582 Japan
| |
Collapse
|
15
|
Narumi R, Tomonaga T. Quantitative Analysis of Tissue Samples by Combining iTRAQ Isobaric Labeling with Selected/Multiple Reaction Monitoring (SRM/MRM). Methods Mol Biol 2016; 1355:85-101. [PMID: 26584920 DOI: 10.1007/978-1-4939-3049-4_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mass spectrometry-based phosphoproteomics is an indispensible technique used in the discovery and quantification of phosphorylation events on proteins in biological samples. The application of this technique to tissue samples is especially useful for the discovery of biomarkers as well as biological studies. We herein describe the application of a large-scale phosphoproteome analysis and SRM/MRM-based quantitation to develop a strategy for the systematic discovery and validation of biomarkers using tissue samples.
Collapse
Affiliation(s)
- Ryohei Narumi
- Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, Osaka, Japan.,Laboratory of Proteome Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Takeshi Tomonaga
- Laboratory of Proteome Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.
| |
Collapse
|
16
|
Nguyen TD, Carrascal M, Vidal-Cortes O, Gallardo O, Casas V, Gay M, Phan VC, Abian J. The phosphoproteome of human Jurkat T cell clones upon costimulation with anti-CD3/anti-CD28 antibodies. J Proteomics 2016; 131:190-198. [DOI: 10.1016/j.jprot.2015.10.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 10/12/2015] [Accepted: 10/28/2015] [Indexed: 10/22/2022]
|
17
|
Kuropka B, Witte A, Sticht J, Waldt N, Majkut P, Hackenberger CPR, Schraven B, Krause E, Kliche S, Freund C. Analysis of Phosphorylation-dependent Protein Interactions of Adhesion and Degranulation Promoting Adaptor Protein (ADAP) Reveals Novel Interaction Partners Required for Chemokine-directed T cell Migration. Mol Cell Proteomics 2015; 14:2961-72. [PMID: 26246585 DOI: 10.1074/mcp.m115.048249] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Indexed: 11/06/2022] Open
Abstract
Stimulation of T cells leads to distinct changes of their adhesive and migratory properties. Signal propagation from activated receptors to integrins depends on scaffolding proteins such as the adhesion and degranulation promoting adaptor protein (ADAP)(1). Here we have comprehensively investigated the phosphotyrosine interactome of ADAP in T cells and define known and novel interaction partners of functional relevance. While most phosphosites reside in unstructured regions of the protein, thereby defining classical SH2 domain interaction sites for master regulators of T cell signaling such as SLP76, Fyn-kinase, and NCK, other binding events depend on structural context. Interaction proteomics using different ADAP constructs comprising most of the known phosphotyrosine motifs as well as the structured domains confirm that a distinct set of proteins is attracted by pY571 of ADAP, including the ζ-chain-associated protein kinase of 70 kDa (ZAP70). The interaction of ADAP and ZAP70 is inducible upon stimulation either of the T cell receptor (TCR) or by chemokine. NMR spectroscopy reveals that the N-terminal SH2 domains within a ZAP70-tandem-SH2 construct is the major site of interaction with phosphorylated ADAP-hSH3(N) and microscale thermophoresis (MST) indicates an intermediate binding affinity (Kd = 2.3 μm). Interestingly, although T cell receptor dependent events such as T cell/antigen presenting cell (APC) conjugate formation and adhesion are not affected by mutation of Y571, migration of T cells along a chemokine gradient is compromised. Thus, although most phospho-sites in ADAP are linked to T cell receptor related functions we have identified a unique phosphotyrosine that is solely required for chemokine induced T cell behavior.
Collapse
Affiliation(s)
- Benno Kuropka
- From the ‡Freie Universität Berlin, Institut für Chemie und Biochemie, Protein Biochemistry group, Thielallee 63, 14195 Berlin, Germany; §Leibniz-Institut für Molekulare Pharmakologie, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Amelie Witte
- ¶Otto-von-Guericke-University, Institute of Molecular and Clinical Immunology, Leipziger Strasse 44, 39120 Magdeburg, Germany
| | - Jana Sticht
- From the ‡Freie Universität Berlin, Institut für Chemie und Biochemie, Protein Biochemistry group, Thielallee 63, 14195 Berlin, Germany
| | - Natalie Waldt
- ¶Otto-von-Guericke-University, Institute of Molecular and Clinical Immunology, Leipziger Strasse 44, 39120 Magdeburg, Germany
| | - Paul Majkut
- §Leibniz-Institut für Molekulare Pharmakologie, Robert-Rössle-Strasse 10, 13125 Berlin, Germany; ‖RiNA GmbH, Volmerstrasse 9, 12489 Berlin, Germany
| | | | - Burkhart Schraven
- ¶Otto-von-Guericke-University, Institute of Molecular and Clinical Immunology, Leipziger Strasse 44, 39120 Magdeburg, Germany; **Helmholtz Center for Infection Research (HZI), Department of Immune Control, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Eberhard Krause
- §Leibniz-Institut für Molekulare Pharmakologie, Robert-Rössle-Strasse 10, 13125 Berlin, Germany;
| | - Stefanie Kliche
- ¶Otto-von-Guericke-University, Institute of Molecular and Clinical Immunology, Leipziger Strasse 44, 39120 Magdeburg, Germany;
| | - Christian Freund
- From the ‡Freie Universität Berlin, Institut für Chemie und Biochemie, Protein Biochemistry group, Thielallee 63, 14195 Berlin, Germany;
| |
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
|
Goodfellow HS, Frushicheva MP, Ji Q, Cheng DA, Kadlecek TA, Cantor AJ, Kuriyan J, Chakraborty AK, Salomon A, Weiss A. The catalytic activity of the kinase ZAP-70 mediates basal signaling and negative feedback of the T cell receptor pathway. Sci Signal 2015; 8:ra49. [PMID: 25990959 DOI: 10.1126/scisignal.2005596] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
T cell activation by antigens binding to the T cell receptor (TCR) must be properly regulated to ensure normal T cell development and effective immune responses to pathogens and transformed cells while avoiding autoimmunity. The Src family kinase Lck and the Syk family kinase ZAP-70 (ζ chain-associated protein kinase of 70 kD) are sequentially activated in response to TCR engagement and serve as critical components of the TCR signaling machinery that leads to T cell activation. We performed a mass spectrometry-based phosphoproteomic study comparing the quantitative differences in the temporal dynamics of phosphorylation in stimulated and unstimulated T cells with or without inhibition of ZAP-70 catalytic activity. The data indicated that the kinase activity of ZAP-70 stimulates negative feedback pathways that target Lck and thereby modulate the phosphorylation patterns of the immunoreceptor tyrosine-based activation motifs (ITAMs) of the CD3 and ζ chain components of the TCR and of signaling molecules downstream of Lck, including ZAP-70. We developed a computational model that provides a mechanistic explanation for the experimental findings on ITAM phosphorylation in wild-type cells, ZAP-70-deficient cells, and cells with inhibited ZAP-70 catalytic activity. This model incorporated negative feedback regulation of Lck activity by the kinase activity of ZAP-70 and predicted the order in which tyrosines in the ITAMs of TCR ζ chains must be phosphorylated to be consistent with the experimental data.
Collapse
Affiliation(s)
- Hanna Sjölin Goodfellow
- Howard Hughes Medical Institute, UCSF, San Francisco, CA 94143, USA.,Department of Medicine, UCSF, San Francisco, CA 94143, USA
| | - Maria P Frushicheva
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Qinqin Ji
- Department of Chemistry, Brown University, Providence, RI 02912, USA
| | - Debra A Cheng
- Howard Hughes Medical Institute, UCSF, San Francisco, CA 94143, USA.,Department of Medicine, UCSF, San Francisco, CA 94143, USA
| | - Theresa A Kadlecek
- Howard Hughes Medical Institute, UCSF, San Francisco, CA 94143, USA.,Department of Medicine, UCSF, San Francisco, CA 94143, USA
| | - Aaron J Cantor
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA.,California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720, USA
| | - John Kuriyan
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA.,California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720, USA.,Department of Chemistry, University of California, Berkeley, CA 94720, USA.,Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA.,Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Arup K Chakraborty
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Arthur Salomon
- Department of Chemistry, Brown University, Providence, RI 02912, USA.,Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Arthur Weiss
- Howard Hughes Medical Institute, UCSF, San Francisco, CA 94143, USA.,Department of Medicine, UCSF, San Francisco, CA 94143, USA
| |
Collapse
|
20
|
Boylan JM, Salomon AR, Tantravahi U, Gruppuso PA. Adaptation of HepG2 cells to a steady-state reduction in the content of protein phosphatase 6 (PP6) catalytic subunit. Exp Cell Res 2015; 335:224-37. [PMID: 25999147 DOI: 10.1016/j.yexcr.2015.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 05/06/2015] [Accepted: 05/08/2015] [Indexed: 11/27/2022]
Abstract
Protein phosphatase 6 (PP6) is a ubiquitous Ser/Thr phosphatase involved in an array of cellular processes. To assess the potential of PP6 as a therapeutic target in liver disorders, we attenuated expression of the PP6 catalytic subunit in HepG2 cells using lentiviral-transduced shRNA. Two PP6 knock-down (PP6KD) cell lines (90% reduction of PP6-C protein content) were studied in depth. Both proliferated at a rate similar to control cells. However, flow cytometry indicated G2/M cell cycle arrest that was accounted for by a shift of the cells from a diploid to tetraploid state. PP6KD cells did not show an increase in apoptosis, nor did they exhibit reduced viability in the presence of bleomycin or taxol. Gene expression analysis by microarray showed attenuated anti-inflammatory signaling. Genes associated with DNA replication were downregulated. Mass spectrometry-based phosphoproteomic analysis yielded 80 phosphopeptides representing 56 proteins that were significantly affected by a stable reduction in PP6-C. Proteins involved in DNA replication, DNA damage repair and pre-mRNA splicing were overrepresented among these. PP6KD cells showed intact mTOR signaling. Our studies demonstrated involvement of PP6 in a diverse set of biological pathways and an adaptive response that may limit the effectiveness of targeting PP6 in liver disorders.
Collapse
Affiliation(s)
- Joan M Boylan
- Department of Pediatrics, Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Arthur R Salomon
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA; Department of Chemistry, Brown University, Providence, RI, USA
| | - Umadevi Tantravahi
- Division of Genetics, Department of Pathology, Brown University and Women and Infants Hospital, Providence, RI, USA
| | - Philip A Gruppuso
- Department of Pediatrics, Brown University and Rhode Island Hospital, Providence, RI, USA; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| |
Collapse
|
21
|
Chylek LA, Wilson BS, Hlavacek WS. Modeling biomolecular site dynamics in immunoreceptor signaling systems. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 844:245-62. [PMID: 25480645 DOI: 10.1007/978-1-4939-2095-2_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The immune system plays a central role in human health. The activities of immune cells, whether defending an organism from disease or triggering a pathological condition such as autoimmunity, are driven by the molecular machinery of cellular signaling systems. Decades of experimentation have elucidated many of the biomolecules and interactions involved in immune signaling and regulation, and recently developed technologies have led to new types of quantitative, systems-level data. To integrate such information and develop nontrivial insights into the immune system, computational modeling is needed, and it is essential for modeling methods to keep pace with experimental advances. In this chapter, we focus on the dynamic, site-specific, and context-dependent nature of interactions in immunoreceptor signaling (i.e., the biomolecular site dynamics of immunoreceptor signaling), the challenges associated with capturing these details in computational models, and how these challenges have been met through use of rule-based modeling approaches.
Collapse
Affiliation(s)
- Lily A Chylek
- Department of Chemistry and Chemical Biology, Cornell University, 14853, Ithaca, NY, USA,
| | | | | |
Collapse
|
22
|
Ji Q, Salomon AR. Wide-scale quantitative phosphoproteomic analysis reveals that cold treatment of T cells closely mimics soluble antibody stimulation. J Proteome Res 2015; 14:2082-9. [PMID: 25839225 DOI: 10.1021/pr501172u] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The activation of T lymphocytes through antigen-mediated T cell receptor (TCR) clustering is vital in regulating the adaptive immune response. Although T cell receptor signaling has been extensively studied, the fundamental mechanisms for signal initiation are not fully understood. Reduced temperatures have initiated some of the hallmarks of TCR signaling, such as increased phosphorylation and activation on ERK and calcium release from the endoplasmic reticulum, as well as coalesced the T cell membrane microdomains. The precise mechanism of the TCR signaling initiation due to temperature change remains obscure. One critical question is whether the signaling initiated by the cold treatment of T cells differs from the signaling initiated by the cross-linking of the T cell receptor. To address this uncertainty, we performed a wide-scale, quantitative mass-spectrometry-based phosphoproteomic analysis on T cells stimulated either by temperature shifts or through the cross-linking of the TCR. Careful statistical comparisons between the two stimulations revealed a striking level of identity among the subset of 339 sites that changed significantly with both stimulations. This study demonstrates for the first time, in unprecedented detail, that T cell cold treatment was sufficient to initiate signaling patterns that were nearly identical to those of soluble antibody stimulation, shedding new light on the mechanism of activation of these critically important immune cells.
Collapse
Affiliation(s)
- Qinqin Ji
- †Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Arthur R Salomon
- †Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States.,‡Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02903, United States
| |
Collapse
|
23
|
Helou YA, Salomon AR. Protein networks and activation of lymphocytes. Curr Opin Immunol 2015; 33:78-85. [PMID: 25687331 DOI: 10.1016/j.coi.2015.01.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/30/2015] [Accepted: 01/30/2015] [Indexed: 12/30/2022]
Abstract
The signal transduction pathways initiated by lymphocyte activation play a critical role in regulating host immunity. High-resolution mass spectrometry has accelerated the investigation of these complex and dynamic pathways by enabling the qualitative and quantitative investigation of thousands of proteins and phosphoproteins simultaneously. In addition, the unbiased and wide-scale identification of protein-protein interaction networks and protein kinase substrates in lymphocyte signaling pathways can be achieved by mass spectrometry-based approaches. Critically, the integration of these discovery-driven strategies with single-cell analysis using mass cytometry can facilitate the understanding of complex signaling phenotypes in distinct immunophenotypes.
Collapse
Affiliation(s)
- Ynes A Helou
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI 02912, USA
| | - Arthur R Salomon
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
24
|
Jouy F, Müller SA, Wagner J, Otto W, von Bergen M, Tomm JM. Integration of conventional quantitative and phospho-proteomics reveals new elements in activated Jurkat T-cell receptor pathway maintenance. Proteomics 2014; 15:25-33. [DOI: 10.1002/pmic.201400119] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 09/23/2014] [Accepted: 10/21/2014] [Indexed: 11/07/2022]
Affiliation(s)
- Florent Jouy
- Department of Proteomics, Helmholtz Centre for Environmental Research - UFZ; Leipzig Germany
| | - Stephan A. Müller
- Department of Proteomics, Helmholtz Centre for Environmental Research - UFZ; Leipzig Germany
| | - Juliane Wagner
- Department of Proteomics, Helmholtz Centre for Environmental Research - UFZ; Leipzig Germany
| | - Wolfgang Otto
- Department of Proteomics, Helmholtz Centre for Environmental Research - UFZ; Leipzig Germany
| | - Martin von Bergen
- Department of Proteomics, Helmholtz Centre for Environmental Research - UFZ; Leipzig Germany
- Department of Metabolomics; Helmholtz Centre for Environmental Research - UFZ; Leipzig Germany
- Department of Biotechnology and Environmental Engineering; University of Aalborg; Aalborg Denmark
| | - Janina M. Tomm
- Department of Proteomics, Helmholtz Centre for Environmental Research - UFZ; Leipzig Germany
| |
Collapse
|
25
|
Ji Q, Ding Y, Salomon AR. SRC homology 2 domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76) N-terminal tyrosine residues regulate a dynamic signaling equilibrium involving feedback of proximal T-cell receptor (TCR) signaling. Mol Cell Proteomics 2014; 14:30-40. [PMID: 25316710 DOI: 10.1074/mcp.m114.037861] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
SRC homology 2 domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76) is a cytosolic adaptor protein that plays an important role in the T-cell receptor-mediated T-cell signaling pathway. SLP-76 links proximal receptor stimulation to downstream effectors through interaction with many signaling proteins. Previous studies showed that mutation of three tyrosine residues, Tyr(112), Tyr(128), and Tyr(145), in the N terminus of SLP-76 results in severely impaired phosphorylation and activation of Itk and PLCγ1, which leads to defective calcium mobilization, Erk activation, and NFAT activation. To expand our knowledge of the role of N-terminal phosphorylation of SLP-76 from these three tyrosine sites, we characterized nearly 1000 tyrosine phosphorylation sites via mass spectrometry in SLP-76 reconstituted wild-type cells and SLP-76 mutant cells in which three tyrosine residues were replaced with phenylalanines (Y3F mutant). Mutation of the three N-terminal tyrosine residues of SLP-76 phenocopied SLP-76-deficient cells for the majority of tyrosine phosphorylation sites observed, including feedback on proximal T-cell receptor signaling proteins. Meanwhile, reversed phosphorylation changes were observed on Tyr(192) of Lck when we compared mutants to the complete removal of SLP-76. In addition, N-terminal tyrosine sites of SLP-76 also perturbed phosphorylation of Tyr(440) of Fyn, Tyr(702) of PLCγ1, Tyr(204), Tyr(397), and Tyr(69) of ZAP-70, revealing new modes of regulation on these sites. All these findings confirmed the central role of N-terminal tyrosine sites of SLP-76 in the pathway and also shed light on novel signaling events that are uniquely regulated by SLP-76 N-terminal tyrosine residues.
Collapse
Affiliation(s)
- Qinqin Ji
- From the ‡Department of Chemistry, Brown University Providence, RI 02903
| | - Yiyuan Ding
- From the ‡Department of Chemistry, Brown University Providence, RI 02903
| | - Arthur R Salomon
- From the ‡Department of Chemistry, Brown University Providence, RI 02903; §Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University Providence, RI 02903
| |
Collapse
|
26
|
Chylek LA, Akimov V, Dengjel J, Rigbolt KTG, Hu B, Hlavacek WS, Blagoev B. Phosphorylation site dynamics of early T-cell receptor signaling. PLoS One 2014; 9:e104240. [PMID: 25147952 PMCID: PMC4141737 DOI: 10.1371/journal.pone.0104240] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 07/07/2014] [Indexed: 11/18/2022] Open
Abstract
In adaptive immune responses, T-cell receptor (TCR) signaling impacts multiple cellular processes and results in T-cell differentiation, proliferation, and cytokine production. Although individual protein-protein interactions and phosphorylation events have been studied extensively, we lack a systems-level understanding of how these components cooperate to control signaling dynamics, especially during the crucial first seconds of stimulation. Here, we used quantitative proteomics to characterize reshaping of the T-cell phosphoproteome in response to TCR/CD28 co-stimulation, and found that diverse dynamic patterns emerge within seconds. We detected phosphorylation dynamics as early as 5 s and observed widespread regulation of key TCR signaling proteins by 30 s. Development of a computational model pointed to the presence of novel regulatory mechanisms controlling phosphorylation of sites with central roles in TCR signaling. The model was used to generate predictions suggesting unexpected roles for the phosphatase PTPN6 (SHP-1) and shortcut recruitment of the actin regulator WAS. Predictions were validated experimentally. This integration of proteomics and modeling illustrates a novel, generalizable framework for solidifying quantitative understanding of a signaling network and for elucidating missing links.
Collapse
Affiliation(s)
- Lily A. Chylek
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
- Center for Nonlinear Studies, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, United States of America
| | - Vyacheslav Akimov
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Jörn Dengjel
- Department of Dermatology, Medical Center; Freiburg Institute for Advanced Studies (FRIAS); BIOSS Centre for Biological Signalling Studies; ZBSA Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany
| | - Kristoffer T. G. Rigbolt
- Department of Dermatology, Medical Center; Freiburg Institute for Advanced Studies (FRIAS); BIOSS Centre for Biological Signalling Studies; ZBSA Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany
| | - Bin Hu
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
- Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - William S. Hlavacek
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
- Center for Nonlinear Studies, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
- Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| |
Collapse
|
27
|
Sjoelund V, Smelkinson M, Nita-Lazar A. Phosphoproteome profiling of the macrophage response to different toll-like receptor ligands identifies differences in global phosphorylation dynamics. J Proteome Res 2014; 13:5185-97. [PMID: 24941444 PMCID: PMC4227906 DOI: 10.1021/pr5002466] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
![]()
Toll-like
receptors (TLRs) are among the first sensors that detect
infection and drive immune response. Macrophages encountering a pathogen
are usually stimulated not by one TLR, but by a combination of TLRs
engaged by distinct microbe ligands. To understand the integrated
signaling under complex conditions, we investigated the differences
in the phosphoprotein signaling cascades triggered by TLR2, TLR4,
and TLR7 ligands using a single responding cell population. We performed
a global, quantitative, early poststimulation kinetic analysis of
the mouse macrophage phosphoproteome using stable isotope labeling
with amino acids coupled to phosphopeptide enrichment and high-resolution
mass spectrometry. For each TLR ligand, we found marked elevation
of phosphorylation of cytoskeleton components, GTPases of the Rho
family, and phospholipase C signaling pathway proteins. Phosphorylation
of proteins involved in phagocytosis was only seen in response to
TLR2 and TLR4 but not to TLR7 activation. Changes in the phosphorylation
of proteins involved in endocytosis were delayed in response to TLR2
as compared to TLR4 ligands. These findings reveal that the phosphoproteomic
response to stimulation of distinct TLRs varies both in the major
modification targets and the phosphorylation dynamics. These results
advance the understanding of how macrophages sense and respond to
a diverse set of TLR stimuli.
Collapse
Affiliation(s)
- Virginie Sjoelund
- Laboratory of Systems Biology, and §Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | | | | |
Collapse
|
28
|
Huang KY, Wu HY, Chen YJ, Lu CT, Su MG, Hsieh YC, Tsai CM, Lin KI, Huang HD, Lee TY, Chen YJ. RegPhos 2.0: an updated resource to explore protein kinase-substrate phosphorylation networks in mammals. Database (Oxford) 2014; 2014:bau034. [PMID: 24771658 PMCID: PMC3999940 DOI: 10.1093/database/bau034] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 03/27/2014] [Accepted: 03/30/2014] [Indexed: 11/13/2022]
Abstract
Protein phosphorylation catalyzed by kinases plays crucial roles in regulating a variety of intracellular processes. Owing to an increasing number of in vivo phosphorylation sites that have been identified by mass spectrometry (MS)-based proteomics, the RegPhos, available online at http://csb.cse.yzu.edu.tw/RegPhos2/, was developed to explore protein phosphorylation networks in human. In this update, we not only enhance the data content in human but also investigate kinase-substrate phosphorylation networks in mouse and rat. The experimentally validated phosphorylation sites as well as their catalytic kinases were extracted from public resources, and MS/MS phosphopeptides were manually curated from research articles. RegPhos 2.0 aims to provide a more comprehensive view of intracellular signaling networks by integrating the information of metabolic pathways and protein-protein interactions. A case study shows that analyzing the phosphoproteome profile of time-dependent cell activation obtained from Liquid chromatography-mass spectrometry (LC-MS/MS) analysis, the RegPhos deciphered not only the consistent scheme in B cell receptor (BCR) signaling pathway but also novel regulatory molecules that may involve in it. With an attempt to help users efficiently identify the candidate biomarkers in cancers, 30 microarray experiments, including 39 cancerous versus normal cells, were analyzed for detecting cancer-specific expressed genes coding for kinases and their substrates. Furthermore, this update features an improved web interface to facilitate convenient access to the exploration of phosphorylation networks for a group of genes/proteins. Database URL: http://csb.cse.yzu.edu.tw/RegPhos2/
Collapse
Affiliation(s)
- Kai-Yao Huang
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320, Taiwan, Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsin-Chu 300, Taiwan and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu 300, Taiwan
| | - Hsin-Yi Wu
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320, Taiwan, Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsin-Chu 300, Taiwan and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu 300, Taiwan
| | - Yi-Ju Chen
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320, Taiwan, Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsin-Chu 300, Taiwan and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu 300, Taiwan
| | - Cheng-Tsung Lu
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320, Taiwan, Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsin-Chu 300, Taiwan and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu 300, Taiwan
| | - Min-Gang Su
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320, Taiwan, Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsin-Chu 300, Taiwan and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu 300, Taiwan
| | - Yun-Chung Hsieh
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320, Taiwan, Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsin-Chu 300, Taiwan and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu 300, Taiwan
| | - Chih-Ming Tsai
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320, Taiwan, Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsin-Chu 300, Taiwan and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu 300, Taiwan
| | - Kuo-I Lin
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320, Taiwan, Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsin-Chu 300, Taiwan and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu 300, Taiwan
| | - Hsien-Da Huang
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320, Taiwan, Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsin-Chu 300, Taiwan and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu 300, Taiwan
| | - Tzong-Yi Lee
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320, Taiwan, Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsin-Chu 300, Taiwan and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu 300, Taiwan
| | - Yu-Ju Chen
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320, Taiwan, Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan, Genomics Research Center, Academia Sinica, Taipei 115, Taiwan, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsin-Chu 300, Taiwan and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu 300, Taiwan
| |
Collapse
|
29
|
DeNardo BD, Holloway MP, Ji Q, Nguyen KT, Cheng Y, Valentine MB, Salomon A, Altura RA. Quantitative phosphoproteomic analysis identifies activation of the RET and IGF-1R/IR signaling pathways in neuroblastoma. PLoS One 2013; 8:e82513. [PMID: 24349301 PMCID: PMC3859635 DOI: 10.1371/journal.pone.0082513] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 10/24/2013] [Indexed: 12/20/2022] Open
Abstract
Neuroblastoma is an embryonal tumor of childhood with a heterogenous clinical presentation that reflects differences in activation of complex biological signaling pathways. Protein phosphorylation is a key component of cellular signal transduction and plays a critical role in processes that control cancer cell growth and survival. We used shotgun LC/MS to compare phosphorylation between a human MYCN amplified neuroblastoma cell line (NB10), modeling a resistant tumor, and a human neural precursor cell line (NPC), modeling a normal baseline neural crest cell. 2181 unique phosphorylation sites representing 1171 proteins and 2598 phosphopeptides were found. Protein kinases accounted for 6% of the proteome, with a predominance of tyrosine kinases, supporting their prominent role in oncogenic signaling pathways. Highly abundant receptor tyrosine kinase (RTK) phosphopeptides in the NB10 cell line relative to the NPC cell line included RET, insulin-like growth factor 1 receptor/insulin receptor (IGF-1R/IR), and fibroblast growth factor receptor 1 (FGFR1). Multiple phosphorylated peptides from downstream mediators of the PI3K/AKT/mTOR and RAS pathways were also highly abundant in NB10 relative to NPC. Our analysis highlights the importance of RET, IGF-1R/IR and FGFR1 as RTKs in neuroblastoma and suggests a methodology that can be used to identify potential novel biological therapeutic targets. Furthermore, application of this previously unexploited technology in the clinic opens the possibility of providing a new wide-scale molecular signature to assess disease progression and prognosis.
Collapse
Affiliation(s)
- Bradley D. DeNardo
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| | - Michael P. Holloway
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| | - Qinqin Ji
- Department of Chemistry, Brown University, Providence, Rhode Island, United States of America
| | - Kevin T. Nguyen
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| | - Yan Cheng
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| | - Marcus B. Valentine
- St. Jude Comprehensive Cancer Center Cytogenetic Shared Resource, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Arthur Salomon
- Department of Molecular and Cellular Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Rachel A. Altura
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
30
|
Tian R. Exploring intercellular signaling by proteomic approaches. Proteomics 2013; 14:498-512. [DOI: 10.1002/pmic.201300259] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 08/15/2013] [Accepted: 08/28/2013] [Indexed: 11/07/2022]
Affiliation(s)
- Ruijun Tian
- Lunenfeld-Tanenbaum Research Institute; Mount Sinai Hospital; Toronto ON Canada
| |
Collapse
|
31
|
Chen Z, Lönnberg T, Lahesmaa R. Holistic systems biology approaches to molecular mechanisms of human helper T cell differentiation to functionally distinct subsets. Scand J Immunol 2013; 78:172-80. [PMID: 23679154 DOI: 10.1111/sji.12071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 05/11/2013] [Indexed: 01/22/2023]
Abstract
Current knowledge of helper T cell differentiation largely relies on data generated from mouse studies. To develop therapeutical strategies combating human diseases, understanding the molecular mechanisms how human naïve T cells differentiate to functionally distinct T helper (Th) subsets as well as studies on human differentiated Th cell subsets is particularly valuable. Systems biology approaches provide a holistic view of the processes of T helper differentiation, enable discovery of new factors and pathways involved and generation of new hypotheses to be tested to improve our understanding of human Th cell differentiation and immune-mediated diseases. Here, we summarize studies where high-throughput systems biology approaches have been exploited to human primary T cells. These studies reveal new factors and signalling pathways influencing T cell differentiation towards distinct subsets, important for immune regulation. Such information provides new insights into T cell biology and into targeting immune system for therapeutic interventions.
Collapse
Affiliation(s)
- Z Chen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | | | | |
Collapse
|
32
|
Roux PP, Thibault P. The coming of age of phosphoproteomics--from large data sets to inference of protein functions. Mol Cell Proteomics 2013; 12:3453-64. [PMID: 24037665 DOI: 10.1074/mcp.r113.032862] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Protein phosphorylation is one of the most common post-translational modifications used in signal transduction to control cell growth, proliferation, and survival in response to both intracellular and extracellular stimuli. This modification is finely coordinated by a network of kinases and phosphatases that recognize unique sequence motifs and/or mediate their functions through scaffold and adaptor proteins. Detailed information on the nature of kinase substrates and site-specific phosphoregulation is required in order for one to better understand their pathophysiological roles. Recent advances in affinity chromatography and mass spectrometry (MS) sensitivity have enabled the large-scale identification and profiling of protein phosphorylation, but appropriate follow-up experiments are required in order to ascertain the functional significance of identified phosphorylation sites. In this review, we present meaningful technical details for MS-based phosphoproteomic analyses and describe important considerations for the selection of model systems and the functional characterization of identified phosphorylation sites.
Collapse
Affiliation(s)
- Philippe P Roux
- Institute for Research in Immunology and Cancer, Université de Montréal, P.O. Box 6128, Station. Centre-ville, Montréal, Québec H3C 3J7, Canada
| | | |
Collapse
|
33
|
Helou YA, Nguyen V, Beik SP, Salomon AR. ERK positive feedback regulates a widespread network of tyrosine phosphorylation sites across canonical T cell signaling and actin cytoskeletal proteins in Jurkat T cells. PLoS One 2013; 8:e69641. [PMID: 23874979 PMCID: PMC3714263 DOI: 10.1371/journal.pone.0069641] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 06/11/2013] [Indexed: 11/18/2022] Open
Abstract
Competing positive and negative signaling feedback pathways play a critical role in tuning the sensitivity of T cell receptor activation by creating an ultrasensitive, bistable switch to selectively enhance responses to foreign ligands while suppressing signals from self peptides. In response to T cell receptor agonist engagement, ERK is activated to positively regulate T cell receptor signaling through phosphorylation of Ser59 Lck. To obtain a wide-scale view of the role of ERK in propagating T cell receptor signaling, a quantitative phosphoproteomic analysis of 322 tyrosine phosphorylation sites by mass spectrometry was performed on the human Jurkat T cell line in the presence of U0126, an inhibitor of ERK activation. Relative to controls, U0126-treated cells showed constitutive decreases in phosphorylation through a T cell receptor stimulation time course on tyrosine residues found on upstream signaling proteins (CD3 chains, Lck, ZAP-70), as well as downstream signaling proteins (VAV1, PLCγ1, Itk, NCK1). Additional constitutive decreases in phosphorylation were found on the majority of identified proteins implicated in the regulation of actin cytoskeleton pathway. Although the majority of identified sites on T cell receptor signaling proteins showed decreases in phosphorylation, Tyr598 of ZAP-70 showed elevated phosphorylation in response to U0126 treatment, suggesting differential regulation of this site via ERK feedback. These findings shed new light on ERK’s role in positive feedback in T cell receptor signaling and reveal novel signaling events that are regulated by this kinase, which may fine tune T cell receptor activation.
Collapse
Affiliation(s)
- Ynes A. Helou
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, Rhode Island, United States of America
| | - Vinh Nguyen
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Samantha P. Beik
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Arthur R. Salomon
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, Rhode Island, United States of America
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, United States of America
- Department of Chemistry, Brown University, Providence, Rhode Island, United States of America
- * E-mail:
| |
Collapse
|
34
|
Liu R, Wang X, Aihara K, Chen L. Early diagnosis of complex diseases by molecular biomarkers, network biomarkers, and dynamical network biomarkers. Med Res Rev 2013; 34:455-78. [PMID: 23775602 DOI: 10.1002/med.21293] [Citation(s) in RCA: 199] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many studies have been carried out for early diagnosis of complex diseases by finding accurate and robust biomarkers specific to respective diseases. In particular, recent rapid advance of high-throughput technologies provides unprecedented rich information to characterize various disease genotypes and phenotypes in a global and also dynamical manner, which significantly accelerates the study of biomarkers from both theoretical and clinical perspectives. Traditionally, molecular biomarkers that distinguish disease samples from normal samples are widely adopted in clinical practices due to their ease of data measurement. However, many of them suffer from low coverage and high false-positive rates or high false-negative rates, which seriously limit their further clinical applications. To overcome those difficulties, network biomarkers (or module biomarkers) attract much attention and also achieve better performance because a network (or subnetwork) is considered to be a more robust form to characterize diseases than individual molecules. But, both molecular biomarkers and network biomarkers mainly distinguish disease samples from normal samples, and they generally cannot ensure to identify predisease samples due to their static nature, thereby lacking ability to early diagnosis. Based on nonlinear dynamical theory and complex network theory, a new concept of dynamical network biomarkers (DNBs, or a dynamical network of biomarkers) has been developed, which is different from traditional static approaches, and the DNB is able to distinguish a predisease state from normal and disease states by even a small number of samples, and therefore has great potential to achieve "real" early diagnosis of complex diseases. In this paper, we comprehensively review the recent advances and developments on molecular biomarkers, network biomarkers, and DNBs in particular, focusing on the biomarkers for early diagnosis of complex diseases considering a small number of samples and high-throughput data (or big data). Detailed comparisons of various types of biomarkers as well as their applications are also discussed.
Collapse
Affiliation(s)
- Rui Liu
- Department of Mathematics, South China University of Technology, Guangzhou, 510640, China
| | | | | | | |
Collapse
|
35
|
Xue L, Wang P, Wang L, Renzi E, Radivojac P, Tang H, Arnold R, Zhu JK, Tao WA. Quantitative measurement of phosphoproteome response to osmotic stress in arabidopsis based on Library-Assisted eXtracted Ion Chromatogram (LAXIC). Mol Cell Proteomics 2013; 12:2354-69. [PMID: 23660473 DOI: 10.1074/mcp.o113.027284] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Global phosphorylation changes in plants in response to environmental stress have been relatively poorly characterized to date. Here we introduce a novel mass spectrometry-based label-free quantitation method that facilitates systematic profiling plant phosphoproteome changes with high efficiency and accuracy. This method employs synthetic peptide libraries tailored specifically as internal standards for complex phosphopeptide samples and accordingly, a local normalization algorithm, LAXIC, which calculates phosphopeptide abundance normalized locally with co-eluting library peptides. Normalization was achieved in a small time frame centered to each phosphopeptide to compensate for the diverse ion suppression effect across retention time. The label-free LAXIC method was further treated with a linear regression function to accurately measure phosphoproteome responses to osmotic stress in Arabidopsis. Among 2027 unique phosphopeptides identified and 1850 quantified phosphopeptides in Arabidopsis samples, 468 regulated phosphopeptides representing 497 phosphosites have shown significant changes. Several known and novel components in the abiotic stress pathway were identified, illustrating the capability of this method to identify critical signaling events among dynamic and complex phosphorylation. Further assessment of those regulated proteins may help shed light on phosphorylation response to osmotic stress in plants.
Collapse
Affiliation(s)
- Liang Xue
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Narumi R, Murakami T, Kuga T, Adachi J, Shiromizu T, Muraoka S, Kume H, Kodera Y, Matsumoto M, Nakayama K, Miyamoto Y, Ishitobi M, Inaji H, Kato K, Tomonaga T. A Strategy for Large-Scale Phosphoproteomics and SRM-Based Validation of Human Breast Cancer Tissue Samples. J Proteome Res 2012; 11:5311-22. [DOI: 10.1021/pr3005474] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ryohei Narumi
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Osaka,
Japan
| | - Tatsuo Murakami
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Osaka,
Japan
| | - Takahisa Kuga
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Osaka,
Japan
| | - Jun Adachi
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Osaka,
Japan
| | - Takashi Shiromizu
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Osaka,
Japan
| | - Satoshi Muraoka
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Osaka,
Japan
| | - Hideaki Kume
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Osaka,
Japan
| | - Yoshio Kodera
- Laboratory of Biomolecular Dynamics, Department of Physics, Kitasato University School of Science, Kanagawa, Japan
- Clinical Proteomics Research Center, Chiba University Hospital, Chiba, Japan
| | - Masaki Matsumoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University Fukuoka, Japan
| | - Keiichi Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University Fukuoka, Japan
| | - Yasuhide Miyamoto
- Department of Immunology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Makoto Ishitobi
- Department
of Breast and Endocrine Surgery, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Hideo Inaji
- Department
of Breast and Endocrine Surgery, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Kikuya Kato
- Research Institute, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Takeshi Tomonaga
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Osaka,
Japan
- Clinical Proteomics Research Center, Chiba University Hospital, Chiba, Japan
| |
Collapse
|
37
|
Cao L, Ding Y, Hung N, Yu K, Ritz A, Raphael BJ, Salomon AR. Quantitative phosphoproteomics reveals SLP-76 dependent regulation of PAG and Src family kinases in T cells. PLoS One 2012; 7:e46725. [PMID: 23071622 PMCID: PMC3469622 DOI: 10.1371/journal.pone.0046725] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 09/07/2012] [Indexed: 12/14/2022] Open
Abstract
The SH2-domain-containing leukocyte protein of 76 kDa (SLP-76) plays a critical scaffolding role in T cell receptor (TCR) signaling. As an adaptor protein that contains multiple protein-binding domains, SLP-76 interacts with many signaling molecules and links proximal receptor stimulation to downstream effectors. The function of SLP-76 in TCR signaling has been widely studied using the Jurkat human leukaemic T cell line through protein disruption or site-directed mutagenesis. However, a wide-scale characterization of SLP-76-dependant phosphorylation events is still lacking. Quantitative profiling of over a hundred tyrosine phosphorylation sites revealed new modes of regulation of phosphorylation of PAG, PI3K, and WASP while reconfirming previously established regulation of Itk, PLCγ, and Erk phosphorylation by SLP-76. The absence of SLP-76 also perturbed the phosphorylation of Src family kinases (SFKs) Lck and Fyn, and subsequently a large number of SFK-regulated signaling molecules. Altogether our data suggests unique modes of regulation of positive and negative feedback pathways in T cells by SLP-76, reconfirming its central role in the pathway.
Collapse
Affiliation(s)
- Lulu Cao
- Department of Chemistry, Brown University, Providence, Rhode Island, United States of America
| | - Yiyuan Ding
- Department of Chemistry, Brown University, Providence, Rhode Island, United States of America
| | - Norris Hung
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Kebing Yu
- Department of Chemistry, Brown University, Providence, Rhode Island, United States of America
| | - Anna Ritz
- Department of Computer Science, Brown University, Providence, Rhode Island, United States of America
| | - Benjamin J. Raphael
- Department of Computer Science, Brown University, Providence, Rhode Island, United States of America
| | - Arthur R. Salomon
- Department of Chemistry, Brown University, Providence, Rhode Island, United States of America
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, United States of America
- * E-mail:
| |
Collapse
|
38
|
Phosphoproteomic analysis of liver homogenates. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2012. [PMID: 22903715 DOI: 10.1007/978-1-61779-959-4_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
Regulation of protein function via reversible phosphorylation is an essential component of cell signaling. Our ability to understand complex phosphorylation networks in the physiological context of a whole organism or tissue remains limited. This is largely due to the technical challenge of isolating serine/threonine phosphorylated peptides from a tissue sample. In the present study, we developed a phosphoproteomic strategy to purify and identify phosphopeptides from a tissue sample by employing protein gel filtration, protein strong anion exchange and strong cation exchange (SCX) chromatography, peptide SCX chromatography, and TiO(2) affinity purification. By applying this strategy to the mass spectrometry-based analysis of rat liver homogenates, we were able to identify with high confidence and quantify over 4,000 unique phosphopeptides. Finally, the reproducibility of our methodology was demonstrated by its application to analysis of the mammalian Target of Rapamycin (mTOR) signaling pathways in liver samples obtained from rats in which hepatic mTOR was activated by refeeding following a period of fasting.
Collapse
|
39
|
Negrutskii B, Vlasenko D, El'skaya A. From global phosphoproteomics to individual proteins: the case of translation elongation factor eEF1A. Expert Rev Proteomics 2012; 9:71-83. [PMID: 22292825 DOI: 10.1586/epr.11.71] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Phosphoproteomics is often aimed at deciphering the modified components of signaling pathways in certain organisms, tissues and pathologies. Phosphorylation of housekeeping proteins, albeit important, usually attracts less attention. Here, we provide targeted analysis of eukaryotic translation elongation factor 1A (eEF1A), which is the main element of peptide elongation machinery. There are 97% homologous A1 and A2 isoforms of eEF1A; their expression in mammalian tissues is mutually exclusive and differentially regulated in development. The A2 isoform reveals proto-oncogenic properties and specifically interacts with some cellular proteins. Several tyrosine residues shown experimentally to be phosphorylated in eEF1A1 are hardly solution accessible, so their phosphorylation could be linked with structural rearrangement of the protein molecule. The possible role of tyrosine phosphorylation in providing the background for structural differences between the 'extended' A1 isoform and the compact oncogenic A2 isoform is discussed. The 'road map' for targeted analysis of any protein of interest using phosphoproteomics data is presented.
Collapse
Affiliation(s)
- Boris Negrutskii
- Institute of Molecular Biology & Genetics, National Academy of Sciences of Ukraine, Kiev, 03680, Ukraine.
| | | | | |
Collapse
|
40
|
Alcolea MP, Casado P, Rodríguez-Prados JC, Vanhaesebroeck B, Cutillas PR. Phosphoproteomic analysis of leukemia cells under basal and drug-treated conditions identifies markers of kinase pathway activation and mechanisms of resistance. Mol Cell Proteomics 2012; 11:453-66. [PMID: 22547687 DOI: 10.1074/mcp.m112.017483] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Protein kinase signaling is fundamental to cell homeostasis and is deregulated in all cancers but varies between patients. Understanding the mechanisms underlying this heterogeneity is critical for personalized targeted therapies. Here, we used a recently established LC-MS/MS platform to profile protein phosphorylation in acute myeloid leukemia cell lines with different sensitivities to kinase inhibitors. The compounds used in this study were originally developed to target Janus kinase, phosphatidylinositol 3-kinase, and MEK. After further validation of the technique, we identified several phosphorylation sites that were inhibited by these compounds but whose intensities did not always correlate with growth inhibition sensitivity. In contrast, several hundred phosphorylation sites that correlated with sensitivity/resistance were not in general inhibited by the compounds. These results indicate that markers of pathway activity may not always be reliable indicators of sensitivity of cancer cells to inhibitors that target such pathways, because the activity of parallel kinases can contribute to resistance. By mining our data we identified protein kinase C isoforms as one of such parallel pathways being more active in resistant cells. Consistent with the view that several parallel kinase pathways were contributing to resistance, inhibitors that target protein kinase C, MEK, and Janus kinase potentiated each other in arresting the proliferation of multidrug-resistant cells. Untargeted/unbiased approaches, such as the one described here, to quantify the activity of the intended target kinase pathway in concert with the activities of parallel kinase pathways will be invaluable to personalize therapies based on kinase inhibitors.
Collapse
Affiliation(s)
- Maria P Alcolea
- Analytical Signalling Group, Centre for Cell Signalling, Barts Cancer Institute, Queen Mary University of London, Barts and the London School of Medicine and Dentistry, London EC1M 6BQ, United Kingdom
| | | | | | | | | |
Collapse
|
41
|
Lichtenfels R, Rappl G, Hombach AA, Recktenwald CV, Dressler SP, Abken H, Seliger B. A proteomic view at T cell costimulation. PLoS One 2012; 7:e32994. [PMID: 22539942 PMCID: PMC3335147 DOI: 10.1371/journal.pone.0032994] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 02/07/2012] [Indexed: 12/31/2022] Open
Abstract
The "two-signal paradigm" in T cell activation predicts that the cooperation of "signal 1," provided by the T cell receptor (TCR) through engagement of major histocompatility complex (MHC)-presented peptide, with "signal 2″ provided by costimulatory molecules, the prototype of which is CD28, is required to induce T cell effector functions. While the individual signalling pathways are well understood, little is known about global changes in the proteome pattern during TCR/CD28-mediated activation. Therefore, comparative 2-DE-based proteome analyses of CD3(+) CD69(-) resting T cells versus cells incubated with (i) the agonistic anti-CD3 antibody OKT3 mimicking signal 1 in absence or presence of IL-2 and/or with (ii) the agonistic antibody 15E8 triggering CD28-mediated signaling were performed. Differentially regulated spots were defined leading to the identification of proteins involved in the regulation of the metabolism, shaping and maintenance of the cytoskeleton and signal transduction. Representative members of the differentially expressed protein families, such as calmodulin (CALM), glyceraldehyde-3-phosphate dehydrogenase (GAPDH), L-lactate dehydrogenase (LDH), Rho GDP-dissociation inhibitor 2 (GDIR2), and platelet basic protein (CXCL7), were independently verified by flow cytometry. Data provide a detailed map of individual protein alterations at the global proteome level in response to TCR/CD28-mediated T cell activation.
Collapse
Affiliation(s)
- Rudolf Lichtenfels
- Institute of Medical Immunology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Gunter Rappl
- Center for Molecular Medicine Cologne (CMMC) and Tumor Genetics Section, Clinic I Internal Medicine, University Cologne, Cologne, Germany
| | - Andreas A. Hombach
- Center for Molecular Medicine Cologne (CMMC) and Tumor Genetics Section, Clinic I Internal Medicine, University Cologne, Cologne, Germany
| | | | - Sven P. Dressler
- Institute of Medical Immunology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Hinrich Abken
- Center for Molecular Medicine Cologne (CMMC) and Tumor Genetics Section, Clinic I Internal Medicine, University Cologne, Cologne, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
42
|
Ruperez P, Gago-Martinez A, Burlingame AL, Oses-Prieto JA. Quantitative phosphoproteomic analysis reveals a role for serine and threonine kinases in the cytoskeletal reorganization in early T cell receptor activation in human primary T cells. Mol Cell Proteomics 2012; 11:171-86. [PMID: 22499768 DOI: 10.1074/mcp.m112.017863] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Protein phosphorylation-dephosphorylation events play a primary role in regulation of almost all aspects of cell function including signal transduction, cell cycle, or apoptosis. Thus far, T cell phosphoproteomics have focused on analysis of phosphotyrosine residues, and little is known about the role of serine/threonine phosphorylation in early activation of the T cell receptor (TCR). Therefore, we performed a quantitative mass spectrometry-based analysis of the global phosphoproteome of human primary T cells in response to 5 min of TCR activation with anti-CD3 antibody. Combining immunoprecipitation with an antiphosphotyrosine antibody, titanium dioxide phosphopeptide enrichment, isobaric tag for the relative and absolute quantitation methodology, and strong cation exchange separation, we were able to identify 2814 phosphopeptides. These unique sites were employed to investigate the site-specific phosphorylation dynamics. Five hundred and seventeen phosphorylation sites showed TCR-responsive changes. We found that upon 5 min of stimulation of the TCR, specific serine and threonine kinase motifs are overrepresented in the set of responsive phosphorylation sites. These phosphorylation events targeted proteins with many different activities and are present in different subcellular locations. Many of these proteins are involved in intracellular signaling cascades related mainly to cytoskeletal reorganization and regulation of small GTPase-mediated signal transduction, probably involved in the formation of the immune synapse.
Collapse
Affiliation(s)
- Patricia Ruperez
- Department of Pharmaceutical Chemistry, Mass Spectrometry Facility, School of Pharmacy, University of California San Francisco, San Francisco, California 94158, USA
| | | | | | | |
Collapse
|
43
|
Tobe BT, Hou J, Crain AM, Singec I, Snyder EY, Brill LM. Phosphoproteomic analysis: an emerging role in deciphering cellular signaling in human embryonic stem cells and their differentiated derivatives. Stem Cell Rev Rep 2012; 8:16-31. [PMID: 22009073 PMCID: PMC3839940 DOI: 10.1007/s12015-011-9317-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cellular signaling is largely controlled by protein phosphorylation. This post-translational modification (PTM) has been extensively analyzed when examining one or a few protein phosphorylation events that effect cell signaling. However, protein kinase-driven signaling networks, comprising total (phospho)proteomes, largely control cell fate. Therefore, large-scale analysis of differentially regulated protein phosphorylation is central to elucidating complex cellular events, including maintenance of pluripotency and differentiation of embryonic stem cells (ESCs). The current technology of choice for total phosphoproteome and combined total proteome plus total phosphoproteome (termed (phospho)proteome) analyses is multidimensional liquid chromatography-(MDLC) tandem mass spectrometry (MS/MS). Advances in the use of MDLC for separation of peptides comprising total (phospho)proteomes, phosphopeptide enrichment, separation of enriched fractions, and quantitative peptide identification by MS/MS have been rapid in recent years, as have improvements in the sensitivity, speed, and accuracy of mass spectrometers. Increasingly deep coverage of (phospho)proteomes is allowing an improved understanding of changes in protein phosphorylation networks as cells respond to stimuli and progress from one undifferentiated or differentiated state to another. Although MDLC-MS/MS studies are powerful, understanding the interpretation of the data is important, and targeted experimental pursuit of biological predictions provided by total (phospho)proteome analyses is needed. (Phospho)proteomic analyses of pluripotent stem cells are in their infancy at this time. However, such studies have already begun to contribute to an improved and accelerated understanding of basic pluripotent stem cell signaling and fate control, especially at the systems-biology level.
Collapse
Affiliation(s)
- Brian T.D. Tobe
- The Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Junjie Hou
- The Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Andrew M. Crain
- The Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Ilyas Singec
- The Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Evan Y. Snyder
- The Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Laurence M. Brill
- The Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| |
Collapse
|
44
|
Dworkin M, Mukherjee S, Jayaprakash C, Das J. Dramatic reduction of dimensionality in large biochemical networks owing to strong pair correlations. J R Soc Interface 2012; 9:1824-35. [PMID: 22378749 DOI: 10.1098/rsif.2011.0896] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Large multi-dimensionality of high-throughput datasets pertaining to cell signalling and gene regulation renders it difficult to extract mechanisms underlying the complex kinetics involving various biochemical compounds (e.g. proteins and lipids). Data-driven models often circumvent this difficulty by using pair correlations of the protein expression levels to produce a small number (fewer than 10) of principal components, each a linear combination of the concentrations, to successfully model how cells respond to different stimuli. However, it is not understood if this reduction is specific to a particular biological system or to nature of the stimuli used in these experiments. We study temporal changes in pair correlations, described by the covariance matrix, between concentrations of different molecular species that evolve following deterministic mass-action kinetics in large biologically relevant reaction networks and show that this dramatic reduction of dimensions (from hundreds to less than five) arises from the strong correlations between different species at any time and is insensitive to the form of the nonlinear interactions, network architecture, and to a wide range of values of rate constants and concentrations. We relate temporal changes in the eigenvalue spectrum of the covariance matrix to low-dimensional, local changes in directions of the system trajectory embedded in much larger dimensions using elementary differential geometry. We illustrate how to extract biologically relevant insights such as identifying significant timescales and groups of correlated chemical species from our analysis. Our work provides for the first time, to our knowledge, a theoretical underpinning for the successful experimental analysis and points to a way to extract mechanisms from large-scale high-throughput datasets.
Collapse
Affiliation(s)
- Michael Dworkin
- Battelle Center for Mathematical Medicine, The Research Institute at the Nationwide Children's Hospital, The Ohio State University, 700 Children's Drive, Columbus, OH 43205, USA
| | | | | | | |
Collapse
|
45
|
Zhong J, Kim MS, Chaerkady R, Wu X, Huang TC, Getnet D, Mitchell CJ, Palapetta SM, Sharma J, O'Meally RN, Cole RN, Yoda A, Moritz A, Loriaux MM, Rush J, Weinstock DM, Tyner JW, Pandey A. TSLP signaling network revealed by SILAC-based phosphoproteomics. Mol Cell Proteomics 2012; 11:M112.017764. [PMID: 22345495 DOI: 10.1074/mcp.m112.017764] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Thymic stromal lymphopoietin (TSLP) is a cytokine that plays diverse roles in the regulation of immune responses. TSLP requires a heterodimeric receptor complex consisting of IL-7 receptor α subunit and its unique TSLP receptor (gene symbol CRLF2) to transmit signals in cells. Abnormal TSLP signaling (e.g. overexpression of TSLP or its unique receptor TSLPR) contributes to the development of a number of diseases including asthma and leukemia. However, a detailed understanding of the signaling pathways activated by TSLP remains elusive. In this study, we performed a global quantitative phosphoproteomic analysis of the TSLP signaling network using stable isotope labeling by amino acids in cell culture. By employing titanium dioxide in addition to antiphosphotyrosine antibodies as enrichment methods, we identified 4164 phosphopeptides on 1670 phosphoproteins. Using stable isotope labeling by amino acids in cell culture-based quantitation, we determined that the phosphorylation status of 226 proteins was modulated by TSLP stimulation. Our analysis identified activation of several members of the Src and Tec families of kinases including Btk, Lyn, and Tec by TSLP for the first time. In addition, we report TSLP-induced phosphorylation of protein phosphatases such as Ptpn6 (SHP-1) and Ptpn11 (Shp2), which has also not been reported previously. Co-immunoprecipitation assays showed that Shp2 binds to the adapter protein Gab2 in a TSLP-dependent manner. This is the first demonstration of an inducible protein complex in TSLP signaling. A kinase inhibitor screen revealed that pharmacological inhibition of PI-3 kinase, Jak family kinases, Src family kinases or Btk suppressed TSLP-dependent cellular proliferation making them candidate therapeutic targets in diseases resulting from aberrant TSLP signaling. Our study is the first phosphoproteomic analysis of the TSLP signaling pathway that greatly expands our understanding of TSLP signaling and provides novel therapeutic targets for TSLP/TSLPR-associated diseases in humans.
Collapse
Affiliation(s)
- Jun Zhong
- McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, 21205 Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
König S, Nimtz M, Scheiter M, Ljunggren HG, Bryceson YT, Jänsch L. Kinome analysis of receptor-induced phosphorylation in human natural killer cells. PLoS One 2012; 7:e29672. [PMID: 22238634 PMCID: PMC3251586 DOI: 10.1371/journal.pone.0029672] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 12/01/2011] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Natural killer (NK) cells contribute to the defense against infected and transformed cells through the engagement of multiple germline-encoded activation receptors. Stimulation of the Fc receptor CD16 alone is sufficient for NK cell activation, whereas other receptors, such as 2B4 (CD244) and DNAM-1 (CD226), act synergistically. After receptor engagement, protein kinases play a major role in signaling networks controlling NK cell effector functions. However, it has not been characterized systematically which of all kinases encoded by the human genome (kinome) are involved in NK cell activation. RESULTS A kinase-selective phosphoproteome approach enabled the determination of 188 kinases expressed in human NK cells. Crosslinking of CD16 as well as 2B4 and DNAM-1 revealed a total of 313 distinct kinase phosphorylation sites on 109 different kinases. Phosphorylation sites on 21 kinases were similarly regulated after engagement of either CD16 or co-engagement of 2B4 and DNAM-1. Among those, increased phosphorylation of FYN, KCC2G (CAMK2), FES, and AAK1, as well as the reduced phosphorylation of MARK2, were reproducibly observed both after engagement of CD16 and co-engagement of 2B4 and DNAM-1. Notably, only one phosphorylation on PAK4 was differentally regulated. CONCLUSIONS The present study has identified a significant portion of the NK cell kinome and defined novel phosphorylation sites in primary lymphocytes. Regulated phosphorylations observed in the early phase of NK cell activation imply these kinases are involved in NK cell signaling. Taken together, this study suggests a largely shared signaling pathway downstream of distinct activation receptors and constitutes a valuable resource for further elucidating the regulation of NK cell effector responses.
Collapse
Affiliation(s)
- Sebastian König
- Department of Molecular Structural Biology, Helmholtz-Zentrum für Infektionsforschung, Braunschweig, Germany
| | - Manfred Nimtz
- Department of Molecular Structural Biology, Helmholtz-Zentrum für Infektionsforschung, Braunschweig, Germany
| | - Maxi Scheiter
- Department of Molecular Structural Biology, Helmholtz-Zentrum für Infektionsforschung, Braunschweig, Germany
| | - Hans-Gustaf Ljunggren
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Yenan T. Bryceson
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Lothar Jänsch
- Department of Molecular Structural Biology, Helmholtz-Zentrum für Infektionsforschung, Braunschweig, Germany
- * E-mail:
| |
Collapse
|
47
|
Affiliation(s)
- Alistair V.G. Edwards
- From the Discipline of Pathology (A.V.G.E., S.J.C.), School of Medical Sciences and School of Molecular Bioscience (S.J.C., M.Y.W.), The University of Sydney, New South Wales, Australia
| | - Stuart J. Cordwell
- From the Discipline of Pathology (A.V.G.E., S.J.C.), School of Medical Sciences and School of Molecular Bioscience (S.J.C., M.Y.W.), The University of Sydney, New South Wales, Australia
| | - Melanie Y. White
- From the Discipline of Pathology (A.V.G.E., S.J.C.), School of Medical Sciences and School of Molecular Bioscience (S.J.C., M.Y.W.), The University of Sydney, New South Wales, Australia
| |
Collapse
|
48
|
Rucevic M, Hixson D, Josic D. Mammalian plasma membrane proteins as potential biomarkers and drug targets. Electrophoresis 2011; 32:1549-64. [PMID: 21706493 DOI: 10.1002/elps.201100212] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Defining the plasma membrane proteome is crucial to understand the role of plasma membrane in fundamental biological processes. Change in membrane proteins is one of the first events that take place under pathological conditions, making plasma membrane proteins a likely source of potential disease biomarkers with prognostic or diagnostic potential. Membrane proteins are also potential targets for monoclonal antibodies and other drugs that block receptors or inhibit enzymes essential to the disease progress. Despite several advanced methods recently developed for the analysis of hydrophobic proteins and proteins with posttranslational modifications, integral membrane proteins are still under-represented in plasma membrane proteome. Recent advances in proteomic investigation of plasma membrane proteins, defining their roles as diagnostic and prognostic disease biomarkers and as target molecules in disease treatment, are presented.
Collapse
Affiliation(s)
- Marijana Rucevic
- COBRE Center for Cancer Research Development, Rhode Island Hospital, Providence, RI, USA
| | | | | |
Collapse
|
49
|
Phosphoproteomic profiling of in vivo signaling in liver by the mammalian target of rapamycin complex 1 (mTORC1). PLoS One 2011; 6:e21729. [PMID: 21738781 PMCID: PMC3125343 DOI: 10.1371/journal.pone.0021729] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 06/07/2011] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Our understanding of signal transduction networks in the physiological context of an organism remains limited, partly due to the technical challenge of identifying serine/threonine phosphorylated peptides from complex tissue samples. In the present study, we focused on signaling through the mammalian target of rapamycin (mTOR) complex 1 (mTORC1), which is at the center of a nutrient- and growth factor-responsive cell signaling network. Though studied extensively, the mechanisms involved in many mTORC1 biological functions remain poorly understood. METHODOLOGY/PRINCIPAL FINDINGS We developed a phosphoproteomic strategy to purify, enrich and identify phosphopeptides from rat liver homogenates. Using the anticancer drug rapamycin, the only known target of which is mTORC1, we characterized signaling in liver from rats in which the complex was maximally activated by refeeding following 48 hr of starvation. Using protein and peptide fractionation methods, TiO(2) affinity purification of phosphopeptides and mass spectrometry, we reproducibly identified and quantified over four thousand phosphopeptides. Along with 5 known rapamycin-sensitive phosphorylation events, we identified 62 new rapamycin-responsive candidate phosphorylation sites. Among these were PRAS40, gephyrin, and AMP kinase 2. We observed similar proportions of increased and reduced phosphorylation in response to rapamycin. Gene ontology analysis revealed over-representation of mTOR pathway components among rapamycin-sensitive phosphopeptide candidates. CONCLUSIONS/SIGNIFICANCE In addition to identifying potential new mTORC1-mediated phosphorylation events, and providing information relevant to the biology of this signaling network, our experimental and analytical approaches indicate the feasibility of large-scale phosphoproteomic profiling of tissue samples to study physiological signaling events in vivo.
Collapse
|
50
|
Radulovic M, Godovac-Zimmermann J. Proteomic approaches to understanding the role of the cytoskeleton in host-defense mechanisms. Expert Rev Proteomics 2011; 8:117-26. [PMID: 21329431 DOI: 10.1586/epr.10.91] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The cytoskeleton is a cellular scaffolding system whose functions include maintenance of cellular shape, enabling cellular migration, division, intracellular transport, signaling and membrane organization. In addition, in immune cells, the cytoskeleton is essential for phagocytosis. Following the advances in proteomics technology over the past two decades, cytoskeleton proteome analysis in resting and activated immune cells has emerged as a possible powerful approach to expand our understanding of cytoskeletal composition and function. However, so far there have only been a handful of studies of the cytoskeleton proteome in immune cells. This article considers promising proteomics strategies that could augment our understanding of the role of the cytoskeleton in host-defense mechanisms.
Collapse
Affiliation(s)
- Marko Radulovic
- Division of Medicine, University College London, 5 University Street, London WC1E 6JF, UK.
| | | |
Collapse
|