1
|
Tsaplina O. Interaction of Serratia proteamaculans with Integrins Activates Invasion-Promoting Signaling Pathways. Int J Mol Sci 2025; 26:3955. [PMID: 40362195 PMCID: PMC12071730 DOI: 10.3390/ijms26093955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
The opportunistic bacteria Serratia proteamaculans are able to penetrate human cells. It was previously shown that the bacterial surface protein OmpX promotes bacterial adhesion. In addition, infection with bacteria that synthesize the OmpX protein enhances the expression of EGFR and β1 integrin involved in the invasion of S. proteamaculans. Therefore, this work was aimed at determining the mechanism of interaction of S. proteamaculans with receptors of eukaryotic cells. Both integrin-linked kinase (ILK) and EGFR tyrosine kinase have been shown to be involved in the invasion of these bacteria. During infection, EGFR is first phosphorylated at Tyr845, which is carried out by c-Src kinase transmitting a signal from nearby receptors. The S. proteamaculans invasion depends on c-Src and focal adhesion kinase (FAK), which can both transmit a signal between β1 integrin and EGFR and participate in cytoskeletal rearrangements. These bacteria have been shown to interact with integrin not through the RGD binding site, and integrin binding to the RGD peptide enhances adhesion, invasion, and expression of α5 and β1 integrin subunits in response to infection. On the other hand, bacterial adhesion and increased expression of integrins during infection are caused by OmpX. Thus, OmpX interacts with integrins, and the participation of the α5 and β1 integrin subunits in the S. proteamaculans invasion allows us to assume that the receptor of OmpX is α5β1 integrin.
Collapse
Affiliation(s)
- Olga Tsaplina
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky av. 4, 194064 St. Petersburg, Russia
| |
Collapse
|
2
|
Chou CL, Jayatissa NU, Kichula ET, Ou SM, Limbutara K, Knepper MA. Phosphoproteomic response to epidermal growth factor in native rat inner medullary collecting duct. Am J Physiol Renal Physiol 2025; 328:F29-F47. [PMID: 39508840 PMCID: PMC11918369 DOI: 10.1152/ajprenal.00182.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/25/2024] [Accepted: 11/01/2024] [Indexed: 12/18/2024] Open
Abstract
Epidermal growth factor (EGF) has important effects in the renal collecting duct to regulate salt and water transport. To identify elements of EGF-mediated signaling in the rat renal inner medullary collecting duct (IMCD), we carried out phosphoproteomic analysis. Biochemically isolated rat IMCD suspensions were treated with 1 µM of EGF or vehicle for 30 min. We performed comprehensive quantitative phosphoproteomics using tandem mass tag (TMT)-labeling of tryptic peptides followed by protein mass spectrometry. We present a data resource reporting all detected phosphorylation sites and their changes in response to EGF. For a total of 29,881 unique phosphorylation sites, 135 sites were increased and 119 sites were decreased based on stringent statistical analysis. The data are provided to users at https://esbl.nhlbi.nih.gov/Databases/EGF-phospho/. The analysis demonstrated that EGF signals through canonical EGF pathways in the renal IMCD. Analysis of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways in which EGF-regulated phosphoproteins are over-represented in native rat IMCD cells confirmed mapping to RAF-MEK-extracellular signal-regulated kinase (ERK) signaling but also pointed to a role for EGF in the regulation of protein translation. A large number of phosphoproteins regulated by EGF contained PDZ domains that are key elements of epithelial polarity determination. We also provide a collecting duct EGF-network map as a user-accessible web resource at https://esbl.nhlbi.nih.gov/Databases/EGF-network/. Overall, the phosphoproteomic data presented provide a useful resource for experimental design and modeling of signaling in the renal collecting duct.NEW & NOTEWORTHY EGF negatively regulates transepithelial water and salt transport across the kidney collecting duct. This study identified phosphoproteins affected by EGF stimulation in normal rat collecting ducts, providing insights into global cell signaling mechanisms. Bioinformatic analyses highlighted enhanced canonical ERK signaling alongside a diminished activity in the PI3K-Akt pathway, which is crucial for cell proliferation and survival. This EGF response differs somewhat from prior studies where both pathways were prominently activated.
Collapse
Affiliation(s)
- Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Nipun U Jayatissa
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Elena T Kichula
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Shuo-Ming Ou
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Kavee Limbutara
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
3
|
He Q, Qu M, Bao H, Xu Y, Shen T, Tan D, Barkat MQ, Xu C, Zeng LH, Wu X. Multiple post-translational modifications ensure EGFR functionality: Potential therapeutic targets to overcome its drug-resistance mutations. Cytokine Growth Factor Rev 2023; 70:41-53. [PMID: 36934069 DOI: 10.1016/j.cytogfr.2023.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
Epidermal growth factor receptor (EGFR) mutation is the most common driver mutation in non-small cell lung cancer (NSCLC). The first-line therapy for advanced NSCLC patients with EGFR-sensitive mutation is the EGFR tyrosine kinase inhibitor (EGFR-TKI). However, most NSCLC patients with EGFR mutation will develop resistant mutations in EGFR-TKI therapy. With further studies, resistance mechanisms represented by EGFR-T790M mutations have revealed the impact of EGFR mutations in situ on EGFR-TKIs sensitivity. The third-generation EGFR-TKIs inhibit both EGFR-sensitive mutations and T790M mutations. The emergence of novel mutations such as EGFR-C797S and EGFR-L718Q may decrease efficacy. Searching for new targets to overcome EGFR-TKI resistance becomes a key challenge. Therefore, an in-depth understanding of the regulatory mechanisms of EGFR is essential to find novel targets to overcome drug-resistant mutations in EGFR-TKIs. EGFR, as a receptor-type tyrosine kinase, undergoes homo/heterodimerization and autophosphorylation upon binding to ligands, which activates multiple downstream signaling pathways. Interestingly, there is growing evidence that the kinase activity of EGFR is affected not only by phosphorylation but also by various post-translational modifications (PTMs, such as S-palmitoylation, S-nitrosylation, Methylation, etc.). In this review, we systematically review the effects of different protein PTMs on EGFR kinase activity and its functionality and suggest that influencing EGFR kinase activity by modulating multiple EGFR sites are potential targets to overcome EGFR-TKIs resistance mutations.
Collapse
Affiliation(s)
- Qiangqiang He
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Meiyu Qu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Pharmacology, Zhejiang University City College, Hangzhou 310015, China
| | - Hangyang Bao
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yana Xu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Tingyu Shen
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Dan Tan
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Muhammad Qasim Barkat
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chengyun Xu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ling-Hui Zeng
- Department of Pharmacology, Zhejiang University City College, Hangzhou 310015, China.
| | - Ximei Wu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
4
|
Swiatnicki MR, Rennhack JP, Ortiz MMO, Hollern DP, Perry AV, Kubiak R, Riveria Riveria SM, O’Reilly S, Andrechek ER. Elevated phosphorylation of EGFR in NSCLC due to mutations in PTPRH. PLoS Genet 2022; 18:e1010362. [PMID: 36054194 PMCID: PMC9477422 DOI: 10.1371/journal.pgen.1010362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 09/15/2022] [Accepted: 07/28/2022] [Indexed: 11/20/2022] Open
Abstract
The role of EGFR in lung cancer is well described with numerous activating mutations that result in phosphorylation and tyrosine kinase inhibitors that target EGFR. While the role of the EGFR kinase in non-small cell lung cancer (NSCLC) is appreciated, control of EGFR signaling pathways through dephosphorylation by phosphatases is not as clear. Through whole genome sequencing we have uncovered conserved V483M Ptprh mutations in PyMT induced tumors. Profiling the downstream events of Ptprh mutant tumors revealed AKT activation, suggesting a key target of PTPRH was EGFR tyrosine 1197. Given the role of EGFR in lung cancer, we explored TCGA data which revealed that a subset of PTPRH mutant tumors shared gene expression profiles with EGFR mutant tumors, but that EGFR mutations and PTPRH mutations were mutually exclusive. Generation of a PTPRH knockout NSCLC cell line resulted in Y1197 phosphorylation of EGFR, and a rescue with expression of wild type PTPRH returned EGFR phosphorylation to parental line values while rescue with catalytically dead PTPRH did not. A dose response curve illustrated that two human NSCLC lines with naturally occurring PTPRH mutations responded to EGFR tyrosine kinase inhibition. Osimertinib treatment of these tumors resulted in a reduction of tumor volume relative to vehicle controls. PTPRH mutation resulted in nuclear pEGFR as seen in immunohistochemistry, suggesting that there may also be a role for EGFR as a transcriptional co-factor. Together these data suggest mutations in PTPRH in NSCLC is inhibitory to PTPRH function, resulting in aberrant EGFR activity and ultimately may result in clinically actionable alterations using existing therapies. One of the major genetic causes of lung cancer is EGFR activity. Traditionally this is caused by mutations in the EGFR receptor tyrosine kinase resulting in unchecked activity, which ultimately results in lung cancer. A series of tyrosine kinase inhibitors have been developed that treat these EGFR positive lung cancers, with remarkable efficacy. Here we describe work from a mouse model that revealed mutations in PTPRH, a phosphatase that we show dephosphorylates EGFR. We show that mutation or loss of wild type PTPRH results in elevated EGFR activity. Searching for similar mutations in human lung cancer revealed that 5% of all lung cancers had PTPRH mutations. Since activation of EGFR by mutation and loss of PTPRH function would be redundant, we tested and demonstrated that these events only occurred separately. Patient data revealed that a subset of PTPRH mutant lung cancer did have elevated EGFR activity. Testing two tumor lines from patients with naturally occurring PTPRH mutations revealed a sensitivity to EGFR inhibitors. The broad implications of this work are that there are a large number of lung cancer patients with PTPRH mutations that could potentially benefit from a revised treatment based on sequencing. Currently the PTPTRH mutations are not detected and these patients are treated with chemotherapy as a standard of care while they could potentially be more effectively treated with EGFR inhibitors. The ability to use EGFR inhibitors in PTPRH mutant lung cancers is a new area for investigation and is the primary impact of this research.
Collapse
Affiliation(s)
- Matthew R. Swiatnicki
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, United States of America
| | - Jonathan P. Rennhack
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States of America
| | - Mylena M. O. Ortiz
- Cell and Molecular Biology, Michigan State University, East Lansing, Michigan, United States of America
| | | | - Ashlee V. Perry
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States of America
| | - Rachel Kubiak
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, United States of America
| | | | - Sandra O’Reilly
- Research Technology Support Facility, Michigan State University, East Lansing, Michigan, United States of America
| | - Eran R. Andrechek
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States of America
- * E-mail:
| |
Collapse
|
5
|
The Mammary Gland: Basic Structure and Molecular Signaling during Development. Int J Mol Sci 2022; 23:ijms23073883. [PMID: 35409243 PMCID: PMC8998991 DOI: 10.3390/ijms23073883] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/22/2022] [Accepted: 03/30/2022] [Indexed: 01/27/2023] Open
Abstract
The mammary gland is a compound, branched tubuloalveolar structure and a major characteristic of mammals. The mammary gland has evolved from epidermal apocrine glands, the skin glands as an accessory reproductive organ to support postnatal survival of offspring by producing milk as a source of nutrition. The mammary gland development begins during embryogenesis as a rudimentary structure that grows into an elementary branched ductal tree and is embedded in one end of a larger mammary fat pad at birth. At the onset of ovarian function at puberty, the rudimentary ductal system undergoes dramatic morphogenetic change with ductal elongation and branching. During pregnancy, the alveolar differentiation and tertiary branching are completed, and during lactation, the mature milk-producing glands eventually develop. The early stages of mammary development are hormonal independent, whereas during puberty and pregnancy, mammary gland development is hormonal dependent. We highlight the current understanding of molecular regulators involved during different stages of mammary gland development.
Collapse
|
6
|
Pellegrini E, Signor L, Singh S, Boeri Erba E, Cusack S. Structures of the inactive and active states of RIP2 kinase inform on the mechanism of activation. PLoS One 2017; 12:e0177161. [PMID: 28545134 PMCID: PMC5436651 DOI: 10.1371/journal.pone.0177161] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/24/2017] [Indexed: 12/14/2022] Open
Abstract
Innate immune receptors NOD1 and NOD2 are activated by bacterial peptidoglycans leading to recruitment of adaptor kinase RIP2, which, upon phosphorylation and ubiquitination, becomes a scaffold for downstream effectors. The kinase domain (RIP2K) is a pharmaceutical target for inflammatory diseases caused by aberrant NOD2-RIP2 signalling. Although structures of active RIP2K in complex with inhibitors have been reported, the mechanism of RIP2K activation remains to be elucidated. Here we analyse RIP2K activation by combining crystal structures of the active and inactive states with mass spectrometric characterization of their phosphorylation profiles. The active state has Helix αC inwardly displaced and the phosphorylated Activation Segment (AS) disordered, whilst in the inactive state Helix αC is outwardly displaced and packed against the helical, non-phosphorylated AS. Biophysical measurements show that the active state is a stable dimer whilst the inactive kinase is in a monomer-dimer equilibrium, consistent with the observed structural differences at the dimer interface. We conclude that RIP2 kinase auto-phosphorylation is intimately coupled to dimerization, similar to the case of BRAF. Our results will help drug design efforts targeting RIP2 as a potential treatment for NOD2-RIP2 related inflammatory diseases.
Collapse
Affiliation(s)
| | - Luca Signor
- University Grenoble Alpes, IBS, Grenoble, France
- CNRS, IBS, Grenoble, France
- CEA, IBS, Grenoble, France
| | - Saurabh Singh
- European Molecular Biology Laboratory, Grenoble, France
| | - Elisabetta Boeri Erba
- University Grenoble Alpes, IBS, Grenoble, France
- CNRS, IBS, Grenoble, France
- CEA, IBS, Grenoble, France
| | - Stephen Cusack
- European Molecular Biology Laboratory, Grenoble, France
- * E-mail:
| |
Collapse
|
7
|
Gonçalves RF, Ferreira MS, de Oliveira DN, Canevarolo R, Achilles MA, D'Ercole DL, Bols PE, Visintin JA, Killian GJ, Catharino RR. Analysis and characterisation of bovine oocyte and embryo biomarkers by matrix-assisted desorption ionisation mass spectrometry imaging. Reprod Fertil Dev 2017; 28:293-301. [PMID: 25228254 DOI: 10.1071/rd14047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 05/22/2014] [Indexed: 12/13/2022] Open
Abstract
In the field of 'single cell analysis', many classical strategies like immunofluorescence and electron microscopy are the primary techniques of choice. However, these methodologies are time consuming and do not permit direct identification of specific molecular classes, such as lipids. In the present study, a novel mass spectrometry-based analytical approach was applied to bovine oocytes and embryos. This new metabolomics-based application uses mass spectrometry imaging (MSI), efficient data processing and multivariate data analysis. Metabolic fingerprinting (MF) was applied to the analysis of unfertilised oocytes, 2-, 4- and 8-cell embryos and blastocysts. A semiquantitative strategy for sphingomyelin [SM (16:0)+Na](+) (m/z 725) and phosphatidylcholine [PC (32:0)+Na](+) (m/z 756) was developed, showing that lipid concentration was useful for selecting the best metabolic biomarkers. This study demonstrates that a combination of MF, MSI features and chemometric analysis can be applied to discriminate cell stages, characterising specific biomarkers and relating them to developmental pathways. This information furthers our understanding of fertilisation and preimplantation events during bovine embryo development.
Collapse
Affiliation(s)
- Roseli F Gonçalves
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, São Paulo University, Av. Prof. Dr. Orlando Marques de Paiva, 87 - Cidade Universitária, 05508-270, São Paulo, SP, Brazil
| | - Mónica S Ferreira
- Innovare Biomarkers Laboratory, Medicine and Experimental Surgery Nucleus, University of Campinas, Rua Cinco de Junho, 350 - Barão Geraldo, 13083-877, Campinas, SP, Brazil
| | - Diogo N de Oliveira
- Innovare Biomarkers Laboratory, Medicine and Experimental Surgery Nucleus, University of Campinas, Rua Cinco de Junho, 350 - Barão Geraldo, 13083-877, Campinas, SP, Brazil
| | - Rafael Canevarolo
- Brazilian Biosciences National Laboratory, National Energy and Material Research Center, Post Office box: 6192, 13083-877, Campinas, SP, Brazil
| | - Marcos A Achilles
- Achilles Genetics Ltda, Rua Padre de Toledo Leite, 20 - Centro, 17400-000, Garça, SP, Brazil
| | - Daniela L D'Ercole
- Achilles Genetics Ltda, Rua Padre de Toledo Leite, 20 - Centro, 17400-000, Garça, SP, Brazil
| | - Peter E Bols
- Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1 Gebouw U 0.09, B-2610, Wilrijk, Belgium
| | - Jose A Visintin
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, São Paulo University, Av. Prof. Dr. Orlando Marques de Paiva, 87 - Cidade Universitária, 05508-270, São Paulo, SP, Brazil
| | - Gary J Killian
- Department of Animal Science, College of Agricultural Sciences, The Pennsylvania State University, 324 Henning Building University Park, PA, 16802, USA
| | - Rodrigo R Catharino
- Innovare Biomarkers Laboratory, Medicine and Experimental Surgery Nucleus, University of Campinas, Rua Cinco de Junho, 350 - Barão Geraldo, 13083-877, Campinas, SP, Brazil
| |
Collapse
|
8
|
Locard-Paulet M, Lim L, Veluscek G, McMahon K, Sinclair J, van Weverwijk A, Worboys JD, Yuan Y, Isacke CM, Jørgensen C. Phosphoproteomic analysis of interacting tumor and endothelial cells identifies regulatory mechanisms of transendothelial migration. Sci Signal 2016; 9:ra15. [PMID: 26861043 PMCID: PMC6485367 DOI: 10.1126/scisignal.aac5820] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The exit of metastasizing tumor cells from the vasculature, extravasation, is regulated by their dynamic interactions with the endothelial cells that line the internal surface of vessels. To elucidate signals controlling tumor cell adhesion to the endothelium and subsequent transendothelial migration, we performed phosphoproteomic analysis to map cell-specific changes in protein phosphorylation that were triggered by contact between metastatic MDA-MB-231 breast cancer cells and endothelial cells. From the 2669 unique phosphorylation sites identified, 77 and 43 were differentially phosphorylated in the tumor cells and endothelial cells, respectively. The receptor tyrosine kinase ephrin type A receptor 2 (EPHA2) exhibited decreased Tyr(772) phosphorylation in the cancer cells upon endothelial contact. Knockdown of EPHA2 increased adhesion of the breast cancer cells to human umbilical vein endothelial cells (HUVECs) and their transendothelial migration in coculture cell assays, as well as early-stage lung colonization in vivo. EPHA2-mediated inhibition of transendothelial migration of breast cancer cells depended on interaction with the ligand ephrinA1 on HUVECs and phosphorylation of EPHA2-Tyr(772). When EPHA2 phosphorylation dynamics were compared between cell lines of different metastatic ability, EPHA2-Tyr(772) was rapidly dephosphorylated after ephrinA1 stimulation specifically in cells targeting the lung. Knockdown of the phosphatase LMW-PTP reduced adhesion and transendothelial migration of the breast cancer cells. Overall, cell-specific phosphoproteomic analysis provides a bidirectional map of contact-initiated signaling between tumor and endothelial cells that can be further investigated to identify mechanisms controlling the transendothelial cell migration of cancer cells.
Collapse
Affiliation(s)
- Marie Locard-Paulet
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK. Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Lindsay Lim
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Giulia Veluscek
- Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Kelly McMahon
- Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - John Sinclair
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Antoinette van Weverwijk
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Jonathan D Worboys
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK. Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Yinyin Yuan
- Centre for Evolution and Cancer and Centre for Molecular Pathology, Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK
| | - Clare M Isacke
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Claus Jørgensen
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK. Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK.
| |
Collapse
|
9
|
Zou L, Cao S, Kang N, Huebert RC, Shah VH. Fibronectin induces endothelial cell migration through β1 integrin and Src-dependent phosphorylation of fibroblast growth factor receptor-1 at tyrosines 653/654 and 766. J Biol Chem 2012; 287:7190-202. [PMID: 22247553 PMCID: PMC3293564 DOI: 10.1074/jbc.m111.304972] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 01/12/2012] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix microenvironment regulates cell phenotype and function. One mechanism by which this is achieved is the transactivation of receptor tyrosine kinases by specific matrix molecules. Here, we demonstrate that the provisional matrix protein, fibronectin (FN), activates fibroblast growth factor (FGF) receptor-1 (FGFR1) independent of FGF ligand in liver endothelial cells. FN activation of FGFR1 requires β1 integrin, as evidenced by neutralizing antibody and siRNA-based studies. Complementary genetic and pharmacologic approaches identify that the non-receptor tyrosine kinase Src is required for FN transactivation of FGFR1. Whereas FGF ligand-induced phosphorylation of FGFR1 preferentially activates ERK, FN-induced phosphorylation of FGFR1 preferentially activates AKT, indicating differential downstream signaling of FGFR1 in response to alternate stimuli. Mutation analysis of known tyrosine residues of FGFR1 reveals that tyrosine 653/654 and 766 residues are required for FN-FGFR1 activation of AKT and chemotaxis. Thus, our study mechanistically dissects a new signaling pathway by which FN achieves endothelial cell chemotaxis, demonstrates how differential phosphorylation profiles of FGFR1 can achieve alternate downstream signals, and, more broadly, highlights the diversity of mechanisms by which the extracellular matrix microenvironment regulates cell behavior through transactivation of receptor tyrosine kinases.
Collapse
Affiliation(s)
- Li Zou
- From the Gastroenterology Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, Minnesota 55905
| | - Sheng Cao
- From the Gastroenterology Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, Minnesota 55905
| | - Ningling Kang
- From the Gastroenterology Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, Minnesota 55905
| | - Robert C. Huebert
- From the Gastroenterology Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, Minnesota 55905
| | - Vijay H. Shah
- From the Gastroenterology Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
10
|
Boeri Erba E, Barylyuk K, Yang Y, Zenobi R. Quantifying Protein–Protein Interactions Within Noncovalent Complexes Using Electrospray Ionization Mass Spectrometry. Anal Chem 2011; 83:9251-9. [DOI: 10.1021/ac201576e] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Elisabetta Boeri Erba
- Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Konstantin Barylyuk
- Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Yang Yang
- Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Renato Zenobi
- Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093 Zurich, Switzerland
| |
Collapse
|
11
|
Díaz ME, González L, Miquet JG, Martínez CS, Sotelo AI, Bartke A, Turyn D. Growth hormone modulation of EGF-induced PI3K-Akt pathway in mice liver. Cell Signal 2011; 24:514-523. [PMID: 22019461 DOI: 10.1016/j.cellsig.2011.10.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 10/07/2011] [Accepted: 10/08/2011] [Indexed: 12/20/2022]
Abstract
The epidermal growth factor (EGF) activates the phosphatidylinositol 3-kinase (PI3K)-Akt cascade among other signaling pathways. This route is involved in cell proliferation and survival, therefore, its dysregulation can promote cancer. Considering the relevance of the PI3K-Akt signaling in cell survival and in the pathogenesis of cancer, and that GH was reported to modulate EGFR expression and signaling, the objective of this study was to analyze the effects of increased GH levels on EGF-induced PI3K-Akt signaling. EGF-induced signaling was evaluated in the liver of GH-overexpressing transgenic mice and in their normal siblings. While Akt expression was increased in GH-overexpressing mice, EGF-induced phosphorylation of Akt, relative to its protein content, was diminished at Ser473 and inhibited at Thr308; consequently, mTOR, which is a substrate of Akt, was not activated by EGF. However, the activation of PDK1, a kinase involved in Akt phosphorylation at Thr308, was not reduced in transgenic mice. Kinetics studies of EGF-induced Akt phosphorylation showed that it is rapidly and transiently induced in GH-overexpressing mice compared with normal siblings. Thus, the expression and activity of phosphatases involved in the termination of the PI3K-Akt signaling were studied. In transgenic mice, neither PTEN nor PP2A were hyperactivated; however, EGF induced the rapid and transient association of SHP-2 to Gab1, which mediates association to EGFR and activation of PI3K. Rapid recruitment of SHP2, which would accelerate the termination of the proliferative signal induced, could be therefore contributing to the diminished EGF-induced activity of Akt in GH-overexpressing mice.
Collapse
Affiliation(s)
- Ma Eugenia Díaz
- Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Facultad de Farmacia y Bioquímica, Junín 956 (1113) Buenos Aires, Argentina
| | - Lorena González
- Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Facultad de Farmacia y Bioquímica, Junín 956 (1113) Buenos Aires, Argentina.
| | - Johanna G Miquet
- Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Facultad de Farmacia y Bioquímica, Junín 956 (1113) Buenos Aires, Argentina
| | - Carolina S Martínez
- Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Facultad de Farmacia y Bioquímica, Junín 956 (1113) Buenos Aires, Argentina
| | - Ana I Sotelo
- Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Facultad de Farmacia y Bioquímica, Junín 956 (1113) Buenos Aires, Argentina
| | - Andrzej Bartke
- Geriatrics Research, Departments of Internal Medicine and Physiology, School of Medicine, Southern Illinois University, Springfield, IL 62794, USA
| | - Daniel Turyn
- Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Facultad de Farmacia y Bioquímica, Junín 956 (1113) Buenos Aires, Argentina
| |
Collapse
|
12
|
β1 integrin controls EGFR signaling and tumorigenic properties of lung cancer cells. Oncogene 2011; 30:4087-96. [DOI: 10.1038/onc.2011.107] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
13
|
Pflieger D, Gonnet F, de la Fuente van Bentem S, Hirt H, de la Fuente A. Linking the proteins--elucidation of proteome-scale networks using mass spectrometry. MASS SPECTROMETRY REVIEWS 2011; 30:268-297. [PMID: 21337599 DOI: 10.1002/mas.20278] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 10/05/2009] [Accepted: 10/05/2009] [Indexed: 05/30/2023]
Abstract
Proteomes are intricate. Typically, thousands of proteins interact through physical association and post-translational modifications (PTMs) to give rise to the emergent functions of cells. Understanding these functions requires one to study proteomes as "systems" rather than collections of individual protein molecules. The abstraction of the interacting proteome to "protein networks" has recently gained much attention, as networks are effective representations, that lose specific molecular details, but provide the ability to see the proteome as a whole. Mostly two aspects of the proteome have been represented by network models: proteome-wide physical protein-protein-binding interactions organized into Protein Interaction Networks (PINs), and proteome-wide PTM relations organized into Protein Signaling Networks (PSNs). Mass spectrometry (MS) techniques have been shown to be essential to reveal both of these aspects on a proteome-wide scale. Techniques such as affinity purification followed by MS have been used to elucidate protein-protein interactions, and MS-based quantitative phosphoproteomics is critical to understand the structure and dynamics of signaling through the proteome. We here review the current state-of-the-art MS-based analytical pipelines for the purpose to characterize proteome-scale networks.
Collapse
Affiliation(s)
- Delphine Pflieger
- Laboratoire Analyse et Modélisation pour la Biologie et l'Environnement, Université d'Evry Val d'Essonne, CNRS UMR 8587, Evry, France
| | | | | | | | | |
Collapse
|
14
|
Zhang G, Fang B, Liu RZ, Lin H, Kinose F, Bai Y, Oguz U, Remily-Wood ER, Li J, Altiok S, Eschrich S, Koomen J, Haura EB. Mass spectrometry mapping of epidermal growth factor receptor phosphorylation related to oncogenic mutations and tyrosine kinase inhibitor sensitivity. J Proteome Res 2011; 10:305-19. [PMID: 21080693 PMCID: PMC3050523 DOI: 10.1021/pr1006203] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The epidermal growth factor receptor (EGFR) plays an important role in cancer by activating downstream signals important in growth and survival. Inhibitors of EGFR are frequently selected as treatment for cancer including lung cancer. We performed an unbiased and comprehensive search for EGFR phosphorylation events related to somatic activating mutations and EGFR inhibitor (erlotinib) sensitivity. EGFR immunoprecipitation combined with high resolution liquid chromatography-mass spectrometry and label free quantitation characterized EGFR phosphorylation. Thirty (30) phosphorylation sites were identified including 12 tyrosine (pY), 12 serine (pS), and 6 threonine (pT). Site-specific phosphorylation was monitored by comparing ion signals from the corresponding unmodified peptide. Phosphorylation sites related to activating mutations in EGFR as well as sensitivity to erlotinib were identified using 31 lung cancer cell lines. We identified three sites (pY1092, pY1110, pY1172) correlated with activating mutations and three sites (pY1110, pY1172, pY1197) correlated with erlotinib sensitivity. Five sites (pT693, pY1092, pY1110, pY1172, and pY1197) were inhibited by erlotinib in concentration-dependent manner. Erlotinib sensitivity was confirmed using liquid chromatography coupled to multiple reaction monitoring (LC-MRM) and quantitative Western blotting. This LC-MS/MS strategy can quantitatively assess site-specific EGFR phosphorylation and can identify relationships between somatic mutations or drug sensitivity and protein phosphorylation.
Collapse
Affiliation(s)
- Guolin Zhang
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
| | - Bin Fang
- Proteomics Core Facility, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
| | - Richard Z. Liu
- Department of Biomedical Informatics, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
| | - Huiyi Lin
- Department of Biostatistics, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
| | - Fumi Kinose
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
| | - Yun Bai
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
| | - Umut Oguz
- Proteomics Core Facility, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
| | - Elizabeth R. Remily-Wood
- Proteomics Core Facility, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
| | - Jiannong Li
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
| | - Soner Altiok
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
| | - Steven Eschrich
- Department of Biomedical Informatics, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
| | - John Koomen
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
- Proteomics Core Facility, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
| | - Eric B. Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute; Tampa, FL, USA 33612
| |
Collapse
|
15
|
Condina MR, Klingler‐Hoffmann M, Hoffmann P. Tyrosine Phosphorylation Enrichment and Subsequent Analysis by MALDI‐TOF/TOF MS/MS and LC‐ESI‐IT‐MS/MS. ACTA ACUST UNITED AC 2010; Chapter 13:Unit13.11. [DOI: 10.1002/0471140864.ps1311s62] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Mark R. Condina
- Adelaide Proteomics Centre, School of Molecular and Biomedical Science, University of Adelaide Adelaide Australia
| | - Manuela Klingler‐Hoffmann
- Chemokine Biology Laboratory, School of Molecular and Biomedical Science, University of Adelaide Adelaide Australia
| | - Peter Hoffmann
- Adelaide Proteomics Centre, School of Molecular and Biomedical Science, University of Adelaide Adelaide Australia
| |
Collapse
|
16
|
Cabodi S, Di Stefano P, Leal MDPC, Tinnirello A, Bisaro B, Morello V, Damiano L, Aramu S, Repetto D, Tornillo G, Defilippi P. Integrins and signal transduction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 674:43-54. [PMID: 20549939 DOI: 10.1007/978-1-4419-6066-5_5] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Integrin signaling has a critical function in organizing cells in tissues during both embryonic development and tissue repair. Following their binding to the extracellular ligands, the intracellular signaling pathways triggered by integrins are directed to two major functions: organization of the actin cytoskeleton and regulation of cell behaviour including survival, differentiation and growth. Basic research conducted in the past twelve years has lead to remarkable breakthroughs in this field. Integrins are catalytically inactive and translate positional cues into biochemical signals by direct and/or functional association with intracellular adaptors, cytosolic tyrosine kinases or growth factor and cytokine receptors. The purpose of this chapter is to highlight recent experimental and conceptual advances in integrin signaling with particular emphasis on the ability of integrins to regulate Fak/Src family kinases (SFKs) activation and the cross-talk with soluble growth factors receptors and cytokines.
Collapse
Affiliation(s)
- Sara Cabodi
- Molecular and Biotechnology Center and Department of Genetics, Biology and Biochemistry, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Galvagni F, Pennacchini S, Salameh A, Rocchigiani M, Neri F, Orlandini M, Petraglia F, Gotta S, Sardone GL, Matteucci G, Terstappen GC, Oliviero S. Endothelial Cell Adhesion to the Extracellular Matrix Induces c-Src–Dependent VEGFR-3 Phosphorylation Without the Activation of the Receptor Intrinsic Kinase Activity. Circ Res 2010; 106:1839-48. [DOI: 10.1161/circresaha.109.206326] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Rationale
:
Integrins cooperate with growth factor receptors to promote downstream signaling for cell proliferation and migration. However, the mechanism of receptor activation is still unknown.
Objective
:
To analyze the mechanism of phosphorylation of the vascular endothelial growth factor receptor (VEGFR)-3 by cell adhesion.
Methods and Results
:
We show that VEGFR-3 phosphorylation, induced by cell attachment to the extracellular matrix, is independent from the intrinsic kinase activity of the receptor, as evidenced from phosphorylation cell adhesion experiments with a mutant kinase dead receptor or in the presence of the specific kinase inhibitor MAZ 51. Cell adhesion experiments in the presence of the c-Src inhibitor PP2 or in fibroblast triple knockout for c-Src, Yes, and Fyn (SYF) demonstrate that VEGFR-3 phosphorylation, induced by extracellular matrix, is mediated by c-Src. Kinase assays in vitro with recombinant c-Src show that VEGFR-3 is a direct c-Src target and mass spectrometry analysis identified the sites phosphorylated by c-Src as tyrosine 830, 833, 853, 1063, 1333, and 1337, demonstrating that integrin-mediated receptor phosphorylation induces a phosphorylation pattern that is distinct from that induced by growth factors. Furthermore, pull-down assays show that integrin-mediated VEGFR-3 phosphorylation activates the recruitment to the receptor of the adaptor proteins CRKI/II and SHC inducing activation of JNK.
Conclusions
:
These data suggest that cell adhesion to extracellular matrix induces a downstream signaling using the tyrosine kinase receptor VEGFR-3 as scaffold.
Collapse
Affiliation(s)
- Federico Galvagni
- From Dipartimento di Biologia Molecolare (F.G., S.P., A.S., M.R., F.N., M.O., S.O.), Università degli Studi di Siena; Dipartimento di Pediatria (F.P.), Ostetricia e Medicina della riproduzione, Università degli Studi di Siena; Siena Biotech (S.G., G.L.S., G.C.T.); and Novartis Vaccines (G.M.), Siena, Italy
| | - Susanna Pennacchini
- From Dipartimento di Biologia Molecolare (F.G., S.P., A.S., M.R., F.N., M.O., S.O.), Università degli Studi di Siena; Dipartimento di Pediatria (F.P.), Ostetricia e Medicina della riproduzione, Università degli Studi di Siena; Siena Biotech (S.G., G.L.S., G.C.T.); and Novartis Vaccines (G.M.), Siena, Italy
| | - Ahmad Salameh
- From Dipartimento di Biologia Molecolare (F.G., S.P., A.S., M.R., F.N., M.O., S.O.), Università degli Studi di Siena; Dipartimento di Pediatria (F.P.), Ostetricia e Medicina della riproduzione, Università degli Studi di Siena; Siena Biotech (S.G., G.L.S., G.C.T.); and Novartis Vaccines (G.M.), Siena, Italy
| | - Marina Rocchigiani
- From Dipartimento di Biologia Molecolare (F.G., S.P., A.S., M.R., F.N., M.O., S.O.), Università degli Studi di Siena; Dipartimento di Pediatria (F.P.), Ostetricia e Medicina della riproduzione, Università degli Studi di Siena; Siena Biotech (S.G., G.L.S., G.C.T.); and Novartis Vaccines (G.M.), Siena, Italy
| | - Francesco Neri
- From Dipartimento di Biologia Molecolare (F.G., S.P., A.S., M.R., F.N., M.O., S.O.), Università degli Studi di Siena; Dipartimento di Pediatria (F.P.), Ostetricia e Medicina della riproduzione, Università degli Studi di Siena; Siena Biotech (S.G., G.L.S., G.C.T.); and Novartis Vaccines (G.M.), Siena, Italy
| | - Maurizio Orlandini
- From Dipartimento di Biologia Molecolare (F.G., S.P., A.S., M.R., F.N., M.O., S.O.), Università degli Studi di Siena; Dipartimento di Pediatria (F.P.), Ostetricia e Medicina della riproduzione, Università degli Studi di Siena; Siena Biotech (S.G., G.L.S., G.C.T.); and Novartis Vaccines (G.M.), Siena, Italy
| | - Felice Petraglia
- From Dipartimento di Biologia Molecolare (F.G., S.P., A.S., M.R., F.N., M.O., S.O.), Università degli Studi di Siena; Dipartimento di Pediatria (F.P.), Ostetricia e Medicina della riproduzione, Università degli Studi di Siena; Siena Biotech (S.G., G.L.S., G.C.T.); and Novartis Vaccines (G.M.), Siena, Italy
| | - Stefano Gotta
- From Dipartimento di Biologia Molecolare (F.G., S.P., A.S., M.R., F.N., M.O., S.O.), Università degli Studi di Siena; Dipartimento di Pediatria (F.P.), Ostetricia e Medicina della riproduzione, Università degli Studi di Siena; Siena Biotech (S.G., G.L.S., G.C.T.); and Novartis Vaccines (G.M.), Siena, Italy
| | - Gian Luca Sardone
- From Dipartimento di Biologia Molecolare (F.G., S.P., A.S., M.R., F.N., M.O., S.O.), Università degli Studi di Siena; Dipartimento di Pediatria (F.P.), Ostetricia e Medicina della riproduzione, Università degli Studi di Siena; Siena Biotech (S.G., G.L.S., G.C.T.); and Novartis Vaccines (G.M.), Siena, Italy
| | - Giacomo Matteucci
- From Dipartimento di Biologia Molecolare (F.G., S.P., A.S., M.R., F.N., M.O., S.O.), Università degli Studi di Siena; Dipartimento di Pediatria (F.P.), Ostetricia e Medicina della riproduzione, Università degli Studi di Siena; Siena Biotech (S.G., G.L.S., G.C.T.); and Novartis Vaccines (G.M.), Siena, Italy
| | - Georg C. Terstappen
- From Dipartimento di Biologia Molecolare (F.G., S.P., A.S., M.R., F.N., M.O., S.O.), Università degli Studi di Siena; Dipartimento di Pediatria (F.P.), Ostetricia e Medicina della riproduzione, Università degli Studi di Siena; Siena Biotech (S.G., G.L.S., G.C.T.); and Novartis Vaccines (G.M.), Siena, Italy
| | - Salvatore Oliviero
- From Dipartimento di Biologia Molecolare (F.G., S.P., A.S., M.R., F.N., M.O., S.O.), Università degli Studi di Siena; Dipartimento di Pediatria (F.P.), Ostetricia e Medicina della riproduzione, Università degli Studi di Siena; Siena Biotech (S.G., G.L.S., G.C.T.); and Novartis Vaccines (G.M.), Siena, Italy
| |
Collapse
|
18
|
ZHU JL, ZHANG K, HE XW, ZHANG YK. New Development of Quantitative Mass Spectrometry-Based Proteomics. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2010. [DOI: 10.3724/sp.j.1096.2010.00434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
19
|
ZHU JL, ZHANG K, HE XW, ZHANG YK. New Developments of Quantitative Mass Spectrometry-based Proteomics. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2010. [DOI: 10.1016/s1872-2040(09)60032-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
20
|
González L, Díaz ME, Miquet JG, Sotelo AI, Fernández D, Dominici FP, Bartke A, Turyn D. GH modulates hepatic epidermal growth factor signaling in the mouse. J Endocrinol 2010; 204:299-309. [PMID: 20032199 PMCID: PMC4208314 DOI: 10.1677/joe-09-0372] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Epidermal growth factor (EGF) is a key regulator of cell survival and proliferation involved in the pathogenesis and progression of different types of cancer. The EGF receptor (EGFR) is activated by binding of the specific ligand but also by transactivation triggered by different growth factors including GH. Chronically, elevated GH levels have been associated with the progression of hepatocellular carcinoma. Considering EGF and GH involvement in cell proliferation and their signaling crosstalk, the objective of the present study was to analyze GH modulatory effects on EGF signaling in liver. For this purpose, GH receptor-knockout (GHR-KO) and GH-overexpressing transgenic mice were used. EGFR content was significantly decreased in GHR-KO mice. Consequently, EGF-induced phosphorylation of EGFR, AKT, ERK1/2, STAT3, and STAT5 was significantly decreased in these mice. In contrast, EGFR content as well as its basal tyrosine phosphorylation was increased in transgenic mice overexpressing GH. However, EGF stimulation caused similar levels of EGFR, AKT, and ERK1/2 phosphorylation in normal and transgenic mice, while EGF induction of STAT3 and STAT5 phosphorylation was inhibited in the transgenic mice. Desensitization of the STATs was related to decreased association of these proteins to the EGFR and increased association between STAT5 and the tyrosine phosphatase SH2-containing phosphatase-2. While GHR knockout is associated with diminished expression of the EGFR and a concomitant decrease in EGF signaling, GH overexpression results in EGFR overexpression with different effects depending on the signaling pathway analyzed: AKT and ERK1/2 pathways are induced by EGF, while STAT3 and STAT5 activation is heterologously desensitized.
Collapse
Affiliation(s)
- Lorena González
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Integrins are cell surface transmembrane receptors that recognize and bind to extracellular matrix proteins and counter receptors. Binding of activated integrins to their ligands induces a vast number of structural and signaling changes within the cell. Large, multimolecular complexes assemble onto the cytoplasmic tails of activated integrins to engage and organize the cytoskeleton, and activate signaling pathways that ultimately lead to changes in gene expression. Additionally, integrin-mediated signaling intersects with growth factor-mediated signaling through various levels of cross-talk. This review discusses recent work that has tremendously broadened our understanding of the complexity of integrin-mediated signaling.
Collapse
|
22
|
Cabodi S, Morello V, Masi A, Cicchi R, Broggio C, Distefano P, Brunelli E, Silengo L, Pavone F, Arcangeli A, Turco E, Tarone G, Moro L, Defilippi P. Convergence of integrins and EGF receptor signaling via PI3K/Akt/FoxO pathway in early gene Egr-1 expression. J Cell Physiol 2008; 218:294-303. [PMID: 18844239 DOI: 10.1002/jcp.21603] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The early gene early growth response (Egr-1), a broadly expressed member of the zing-finger family of transcription factors, is induced in many cell types by a variety of growth and differentiation stimuli, including epidermal growth factor (EGF). Here we demonstrate that Egr-1 expression is mainly regulated by integrin-mediated adhesion. Integrin-dependent adhesion plays a dual role in Egr-1 regulation, either being sufficient "per se" to induce Egr-1, or required for EGF-dependent expression of Egr-1, which occurs only in adherent cells and not in cells in suspension. To dissect the molecular basis of integrin-dependent Egr-1 regulation, we show by FLIM-based FRET that in living cells beta1-integrin associates with the EGF receptor (EGFR) and that EGF further increases the extent complex formation. Interestingly, Egr-1 induction depends on integrin-dependent PI3K/Akt activation, as indicated by the decrease in Egr-1 levels in presence of the pharmacological inhibitor LY294002, the kinase-defective Akt mutant and Akt1/2 shRNAs. Indeed, upon adhesion activated Akt translocates into the nucleus and phosphorylates FoxO1, a Forkhead transcription factors. Consistently, FoxO1silencing results in Egr-1-increased levels, indicating that FoxO1 behaves as a negative regulator of Egr-1 expression. These data demonstrate that integrin/EGFR cross-talk is required for expression of Egr-1 through a novel regulatory cascade involving the activation of the PI3K/Akt/Forkhead pathway.
Collapse
Affiliation(s)
- Sara Cabodi
- Centro di Biotecnologie Molecolari and Dipartimento di Genetica, Biologia e Biochimica, Università di Torino, Torino, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Schreiber TB, Mäusbacher N, Breitkopf SB, Grundner-Culemann K, Daub H. Quantitative phosphoproteomics--an emerging key technology in signal-transduction research. Proteomics 2008; 8:4416-32. [PMID: 18837465 DOI: 10.1002/pmic.200800132] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Protein phosphorylation is the most important type of reversible post-translational modification involved in the regulation of cellular signal-transduction processes. In addition to controlling normal cellular physiology on the molecular level, perturbations of phosphorylation-based signaling networks and cascades have been implicated in the onset and progression of various human diseases. Recent advances in mass spectrometry-based proteomics helped to overcome many of the previous limitations in protein phosphorylation analysis. Improved isotope labeling and phosphopeptide enrichment strategies in conjunction with more powerful mass spectrometers and advances in data analysis have been integrated in highly efficient phosphoproteomics workflows, which are capable of monitoring up to several thousands of site-specific phosphorylation events within one large-scale analysis. Combined with ongoing efforts to define kinase-substrate relationships in intact cells, these major achievements have considerable potential to assess phosphorylation-based signaling networks on a system-wide scale. Here, we provide an overview of these exciting developments and their potential to transform signal-transduction research into a technology-driven, high-throughput science.
Collapse
Affiliation(s)
- Thiemo B Schreiber
- Department of Molecular Biology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | | | | | | |
Collapse
|
24
|
Atsriku C, Britton DJ, Held JM, Schilling B, Scott GK, Gibson BW, Benz CC, Baldwin MA. Systematic mapping of posttranslational modifications in human estrogen receptor-alpha with emphasis on novel phosphorylation sites. Mol Cell Proteomics 2008; 8:467-80. [PMID: 18984578 DOI: 10.1074/mcp.m800282-mcp200] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A systematic study of posttranslational modifications of the estrogen receptor isolated from the MCF-7 human breast cancer cell line is reported. Proteolysis with multiple enzymes, mass spectrometry, and tandem mass spectrometry achieved very high sequence coverage for the full-length 66-kDa endogenous protein from estradiol-treated cell cultures. Nine phosphorylated serine residues were identified, three of which were previously unreported and none of which were previously observed by mass spectrometry by any other laboratory. Two additional modified serine residues were identified in recombinant protein, one previously reported but not observed here in endogenous protein and the other previously unknown. Although major emphasis was placed on identifying new phosphorylation sites, N-terminal loss of methionine accompanied by amino acetylation and a lysine side chain acetylation (or possibly trimethylation) were also detected. The use of both HPLC-ESI and MALDI interfaced to different mass analyzers gave higher sequence coverage and identified more sites than could be achieved by either method alone. The estrogen receptor is critical in the development and progression of breast cancer. One previously unreported phosphorylation site identified here was shown to be strongly dependent on estradiol, confirming its potential significance to breast cancer. Greater knowledge of this array of posttranslational modifications of estrogen receptor, particularly phosphorylation, will increase our understanding of the processes that lead to estradiol-induced activation of this protein and may aid the development of therapeutic strategies for management of hormone-dependent breast cancer.
Collapse
|
25
|
Li X, Huang Y, Jiang J, Frank SJ. ERK-dependent threonine phosphorylation of EGF receptor modulates receptor downregulation and signaling. Cell Signal 2008; 20:2145-2155. [PMID: 18762250 PMCID: PMC2613789 DOI: 10.1016/j.cellsig.2008.08.006] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 08/03/2008] [Accepted: 08/11/2008] [Indexed: 10/21/2022]
Abstract
Epidermal growth factor (EGF) signaling is critical in normal and aberrant cellular behavior. Extracellular signal-regulated kinase (ERK) mediates important downstream aspects of EGF signaling. Additionally, EGFR undergoes MEK1-dependent ERK consensus site phosphorylation in response to EGF or cytokines such as growth hormone (GH) and prolactin (PRL). GH- or PRL-induced EGFR phosphorylation alters subsequent EGF-induced EGFR downregulation and signal characteristics in an ERK-dependent fashion. We now use reconstitution to study mutation of the sole EGFR ERK phosphorylation consensus residue, (669)T. CHO-GHR cells, which lack EGFR and express GHR, were stably transfected to express human wild-type or T669A ((669)T changed to alanine) EGFRs at similar abundance. Treatment of cells with GH or EGF caused phosphorylation of WT, but not T669A EGFR, in an ERK activity-dependent fashion that was detected with an antibody that recognizes phosphorylation of ERK consensus sites, indicating that (669)T is required for this phosphorylation. Notably, EGF-induced downregulation of EGFR abundance was much more rapid in cells expressing EGFR T669A vs. WT EGFR. Further, pretreatment with the MEK1/ERK inhibitor PD98059 enhanced EGF-induced EGFR loss in cells expressing WT EGFR, but not EGFR T669A, suggesting that the ERK-dependent effects on EGFR downregulation required phosphorylation of (669)T. In signaling experiments, EGFR T669A displayed enhanced acute (15 min) EGFR tyrosine phosphorylation (reflecting EGFR kinase activity) compared to WT EGFR. Further, acute EGF-induced ubiquitination of WT EGFR was markedly enhanced by PD98059 pretreatment and was increased in EGFR T669A-expressing cells independent of PD98059. These signaling data suggest that ERK-mediated (669)T phosphorylation negatively modulates EGF-induced EGFR kinase activity. We furthered these investigations using a human fibrosarcoma cell line that endogenously expresses EGFR and ErbB-2 and also harbors an activating Ras mutation. In these cells, EGFR was constitutively detected with the ERK consensus site phosphorylation-specific antibody and EGF-induced EGFR downregulation was modest, but was substantially enhanced by pretreatment with MEK1/ERK inhibitor. Collectively, these data indicate that ERK activity, by phosphorylation of a threonine residue in the EGFR juxtamembrane cytoplasmic domain, modulates EGFR trafficking and signaling.
Collapse
Affiliation(s)
- Xin Li
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0012
| | - Yao Huang
- Department of Obstetrics and Gynecology, St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85004
| | - Jing Jiang
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294-0012
| | - Stuart J. Frank
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294-0012
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294-0012
- Endocrinology Section, Medical Service, Veterans Affairs Medical Center, Birmingham, AL 35233
| |
Collapse
|
26
|
Morandell S, Stasyk T, Skvortsov S, Ascher S, Huber LA. Quantitative proteomics and phosphoproteomics reveal novel insights into complexity and dynamics of the EGFR signaling network. Proteomics 2008; 8:4383-401. [DOI: 10.1002/pmic.200800204] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
27
|
Smith JC, Figeys D. Recent developments in mass spectrometry-based quantitative phosphoproteomicsThis paper is one of a selection of papers published in this Special Issue, entitled CSBMCB — Systems and Chemical Biology, and has undergone the Journal's usual peer review process. Biochem Cell Biol 2008; 86:137-48. [DOI: 10.1139/o08-007] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Protein phosphorylation is a reversible post-translational modification that is involved in virtually all eukaryotic cellular processes and has been studied in great detail in recent years. Many developments in mass spectrometry (MS)-based proteomics have been successfully applied to study protein phosphorylation in highly complicated samples. Furthermore, the emergence of a variety of enrichment strategies has allowed some of the challenges associated with low phosphorylation stoichiometry and phosphopeptide copy number to be overcome. The dynamic nature of protein phosphorylation complicates its analysis; however, a number of methods have been developed to successfully quantitate phosphorylation changes in a variety of cellular systems. The following review details some of the most recent breakthroughs in the study of protein phosphorylation, or phosphoproteomics, using MS-based approaches. The majority of the focus is placed on detailing strategies that are currently used to conduct MS-based quantitative phosphoproteomics.
Collapse
Affiliation(s)
- Jeffrey C. Smith
- Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Daniel Figeys
- Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
28
|
Frank SJ. Mechanistic aspects of crosstalk between GH and PRL and ErbB receptor family signaling. J Mammary Gland Biol Neoplasia 2008; 13:119-29. [PMID: 18236142 DOI: 10.1007/s10911-008-9065-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Accepted: 01/02/2008] [Indexed: 11/27/2022] Open
Abstract
Growth hormone (GH) and prolactin (PRL) are anterior pituitary hormones that have multiple roles in growth and metabolism. Both hormones are important in mammary development and breast cancer. The epidermal growth factor (EGF) family of peptides and the receptors that they activate (the ErbB family) are also major players in mammary biology and pathophysiology. Recent studies in signal transduction have highlighted the interplay between signaling pathways referred to as crosstalk. In this review, cell biological and signaling studies related to crosstalk between GH and PRL and the ErbB family are discussed. In particular, the role of GH- and PRL-induced phosphorylation of ErbB receptors in regulating EGF responsiveness is highlighted with attention to potential pathophysiological relevance.
Collapse
Affiliation(s)
- Stuart J Frank
- Department of Cell Biology and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294-0012, USA.
| |
Collapse
|
29
|
Tong J, Taylor P, Jovceva E, St-Germain JR, Jin LL, Nikolic A, Gu X, Li ZH, Trudel S, Moran MF. Tandem Immunoprecipitation of Phosphotyrosine-Mass Spectrometry (TIPY-MS) Indicates C19ORF19 Becomes Tyrosine-Phosphorylated and Associated with Activated Epidermal Growth Factor Receptor. J Proteome Res 2008; 7:1067-77. [DOI: 10.1021/pr7006363] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Jiefei Tong
- Program in Molecular Structure and Function, Hospital for Sick Children, Department of Molecular Genetics, and Banting & Best Department of Medical Research, University of Toronto, Toronto, Canada, McLaughlin Centre for Molecular Medicine, Toronto, Canada, and Hematology-Oncology, Princess Margaret Hospital, Toronto, Canada
| | - Paul Taylor
- Program in Molecular Structure and Function, Hospital for Sick Children, Department of Molecular Genetics, and Banting & Best Department of Medical Research, University of Toronto, Toronto, Canada, McLaughlin Centre for Molecular Medicine, Toronto, Canada, and Hematology-Oncology, Princess Margaret Hospital, Toronto, Canada
| | - Eleonora Jovceva
- Program in Molecular Structure and Function, Hospital for Sick Children, Department of Molecular Genetics, and Banting & Best Department of Medical Research, University of Toronto, Toronto, Canada, McLaughlin Centre for Molecular Medicine, Toronto, Canada, and Hematology-Oncology, Princess Margaret Hospital, Toronto, Canada
| | - Jonathan R. St-Germain
- Program in Molecular Structure and Function, Hospital for Sick Children, Department of Molecular Genetics, and Banting & Best Department of Medical Research, University of Toronto, Toronto, Canada, McLaughlin Centre for Molecular Medicine, Toronto, Canada, and Hematology-Oncology, Princess Margaret Hospital, Toronto, Canada
| | - Lily L. Jin
- Program in Molecular Structure and Function, Hospital for Sick Children, Department of Molecular Genetics, and Banting & Best Department of Medical Research, University of Toronto, Toronto, Canada, McLaughlin Centre for Molecular Medicine, Toronto, Canada, and Hematology-Oncology, Princess Margaret Hospital, Toronto, Canada
| | - Ana Nikolic
- Program in Molecular Structure and Function, Hospital for Sick Children, Department of Molecular Genetics, and Banting & Best Department of Medical Research, University of Toronto, Toronto, Canada, McLaughlin Centre for Molecular Medicine, Toronto, Canada, and Hematology-Oncology, Princess Margaret Hospital, Toronto, Canada
| | - Xiaoping Gu
- Program in Molecular Structure and Function, Hospital for Sick Children, Department of Molecular Genetics, and Banting & Best Department of Medical Research, University of Toronto, Toronto, Canada, McLaughlin Centre for Molecular Medicine, Toronto, Canada, and Hematology-Oncology, Princess Margaret Hospital, Toronto, Canada
| | - Zhi Hua Li
- Program in Molecular Structure and Function, Hospital for Sick Children, Department of Molecular Genetics, and Banting & Best Department of Medical Research, University of Toronto, Toronto, Canada, McLaughlin Centre for Molecular Medicine, Toronto, Canada, and Hematology-Oncology, Princess Margaret Hospital, Toronto, Canada
| | - Suzanne Trudel
- Program in Molecular Structure and Function, Hospital for Sick Children, Department of Molecular Genetics, and Banting & Best Department of Medical Research, University of Toronto, Toronto, Canada, McLaughlin Centre for Molecular Medicine, Toronto, Canada, and Hematology-Oncology, Princess Margaret Hospital, Toronto, Canada
| | - Michael F. Moran
- Program in Molecular Structure and Function, Hospital for Sick Children, Department of Molecular Genetics, and Banting & Best Department of Medical Research, University of Toronto, Toronto, Canada, McLaughlin Centre for Molecular Medicine, Toronto, Canada, and Hematology-Oncology, Princess Margaret Hospital, Toronto, Canada
| |
Collapse
|
30
|
Schuchardt S, Borlak J. Quantitative mass spectrometry to investigate epidermal growth factor receptor phosphorylation dynamics. MASS SPECTROMETRY REVIEWS 2008; 27:51-65. [PMID: 18023079 DOI: 10.1002/mas.20155] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Identifying proteins of signaling networks has received much attention, because an array of biological processes are entirely dependent on protein cross-talk and protein-protein interactions. Protein posttranslational modifications (PTM) add an additional layer of complexity, resulting in complex signaling networks. Of particular interest to our working group are the signaling networks of epidermal growth factor (EGF) receptor, a transmembrane receptor tyrosine kinase involved in various cellular processes, including cell proliferation, differentiation, and survival. Ligand binding to the N-terminal residue of the extracellular domain of EGF receptor induces conformational changes, dimerization, and (auto)-phosphorylation of intracellular tyrosine residues. In addition, activated EGF receptor may positively affect survival pathways, and thus determines the pathways for tumor growth and progression. Notably, in many human malignancies exaggerated EGF receptor activities are commonly observed. An understanding of the mechanism that results in aberrant phosphorylation of EGF receptor tyrosine residues and derived signaling cascades is crucial for an understanding of molecular mechanisms in cancer development. Here, we summarize recent labeling methods and discuss the difficulties in quantitative MS-based phosphorylation assays to probe for receptor tyrosine kinase (RTK) activity. We also review recent advances in sample preparation to investigate membrane-bound RTKs, MS-based detection of phosphopeptides, and the diligent use of different quantitative methods for protein labeling.
Collapse
Affiliation(s)
- Sven Schuchardt
- Department of Drug Research and Medical Biotechnology, Fraunhofer Institute of Toxicology and Experimental Medicine ITEM, Nikolai-Fuchs-Strasse 1, Hannover, Germany
| | | |
Collapse
|
31
|
Liang X, Fonnum G, Hajivandi M, Stene T, Kjus NH, Ragnhildstveit E, Amshey JW, Predki P, Pope RM. Quantitative comparison of IMAC and TiO2 surfaces used in the study of regulated, dynamic protein phosphorylation. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2007; 18:1932-44. [PMID: 17870612 DOI: 10.1016/j.jasms.2007.08.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2007] [Revised: 07/31/2007] [Accepted: 08/03/2007] [Indexed: 05/17/2023]
Abstract
Protein phosphorylation regulates many aspects of cellular function, including cell proliferation, migration, and signal transduction. An efficient strategy to isolate phosphopeptides from a pool of unphosphorylated peptides is essential to global characterization using mass spectrometry. We describe an approach employing isotope tagging reagents for relative and absolute quantification (iTRAQ) labeling to compare quantitatively commercial and prototypal immobilized metal affinity chelate (IMAC) and metal oxide resins. Results indicate a prototype iron chelate resin coupled to magnetic beads outperforms either the Ga(3+)-coupled analog, Fe(3+), or Ga(3+)-loaded, iminodiacetic acid (IDA)-coated magnetic particles, Ga(3+)-loaded Captivate beads, Fe(3+)-loaded Poros 20MC, or zirconium-coated ProteoExtract magnetic beads. For example, compared with Poros 20MC, the magnetic metal chelate (MMC) studied here improved phosphopeptide recovery by 20% and exhibited 60% less contamination from unphosphorylated peptides. With respect to efficiency and contamination, MMC performed as well as prototypal magnetic metal oxide-coated (TiO(2)) beads (MMO) or TiO(2) chromatographic spheres, even if the latter were used with 2,5-dihydroxybenzoic acid (DHB) procedures. Thus far, the sensitivity of the new prototypes reaches 50 fmol, which is comparable to TiO(2) spheres. In an exploration of natural proteomes, tryptic (phospho)peptides captured from stable isotopic labeling with amino acids in cell culture (SILAC)-labeled immunocomplexes following EGF-treatment of 5 x 10(7) HeLa cells were sufficient to quantify stimulated response of over 60 proteins and identify 20 specific phosphorylation sites.
Collapse
Affiliation(s)
- Xiquan Liang
- Invitrogen Corporation, Carlsbad, California, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Zhang X, Ye J, Jensen ON, Roepstorff P. Highly Efficient Phosphopeptide Enrichment by Calcium Phosphate Precipitation Combined with Subsequent IMAC Enrichment. Mol Cell Proteomics 2007; 6:2032-42. [PMID: 17675664 DOI: 10.1074/mcp.m700278-mcp200] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
A new method for enrichment of phosphopeptides in complex mixtures derived by proteolytic digestion of biological samples has been developed. The method is based on calcium phosphate precipitation of the phosphopeptides prior to further enrichment with established affinity enrichment methods. Calcium phosphate precipitation combined with phosphopeptide enrichment using Fe(III) IMAC provided highly selective enrichment of phosphopeptides. Application of the method to a complex peptide sample derived from rice embryo resulted in more than 90% phosphopeptides in the enriched sample as determined by mass spectrometry. Introduction of a two-step IMAC enrichment procedure after calcium phosphate precipitation resulted in observation of an increased number of phosphopeptides.
Collapse
Affiliation(s)
- Xumin Zhang
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark
| | | | | | | |
Collapse
|
33
|
Nakamura JL. The epidermal growth factor receptor in malignant gliomas: pathogenesis and therapeutic implications. Expert Opin Ther Targets 2007; 11:463-72. [PMID: 17373877 DOI: 10.1517/14728222.11.4.463] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Activated epidermal growth factor receptor (EGFR) has emerged as an important therapeutic target for a variety of solid tumors, particularly malignant gliomas. Mutation or amplification of EGFR is commonly observed in malignant gliomas and these modifications are associated with increased cell proliferation and radiation resistance. Small-molecule kinase inhibitors targeting the intracellular kinase domain of the EGFR and monoclonal antibodies against the extracellular domain of the EGFR have demonstrated in vitro efficacy and have spawned clinical trials incorporating EGFR inhibition into the management of malignant gliomas, for example, combining EGFR inhibitors with radiation therapy. This early clinical experience indicates that EGFR inhibitors are well tolerated; however, it remains unclear how best to integrate EGFR inhibition into the management of malignant gliomas. As signaling pathways become better defined, patients may be treated with EGFR inhibitors based on the molecular features of their tumors and treatment efficacy may be improved by combining EGFR inhibition with other small kinase inhibitors and radiation therapy.
Collapse
Affiliation(s)
- Jean L Nakamura
- University of California, San Francisco, Department of Radiation Oncology, 1600 Divisadero Street, Suite H1031, San Francisco, CA 94143, USA.
| |
Collapse
|
34
|
Wissing J, Jänsch L, Nimtz M, Dieterich G, Hornberger R, Kéri G, Wehland J, Daub H. Proteomics Analysis of Protein Kinases by Target Class-selective Prefractionation and Tandem Mass Spectrometry. Mol Cell Proteomics 2007; 6:537-47. [PMID: 17192257 DOI: 10.1074/mcp.t600062-mcp200] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein kinases constitute a large superfamily of enzymes with key regulatory functions in nearly all signal transmission processes of eukaryotic cells. However, due to their relatively low abundance compared with the vast majority of cellular proteins, currently available proteomics techniques do not permit the comprehensive biochemical characterization of protein kinases. To address these limitations, we have developed a prefractionation strategy that uses a combination of immobilized low molecular weight inhibitors for the selective affinity capture of protein kinases. This approach resulted in the direct purification of cell type-specific sets of expressed protein kinases, and more than 140 different members of this enzyme family could be detected by LC-MS/MS. Furthermore the enrichment technique combined with phosphopeptide fractionation led to the identification of more than 200 different phosphorylation sites on protein kinases, which often remain occluded in global phosphoproteome analysis. As the phosphorylation states of protein kinases can provide a readout for the signaling activities within a cellular system, kinase-selective phosphoproteomics based on the procedures described here has the potential to become an important tool in signal transduction analysis.
Collapse
Affiliation(s)
- Josef Wissing
- Department of Cell Biology, Helmholtz Centre for Infection Research (HZI), Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Tan F, Zhang Y, Wang J, Wei J, Qin P, Cai Y, Qian X. Specific capture of phosphopeptides on matrix-assisted laser desorption/ionization time-of-flight mass spectrometry targets modified by magnetic affinity nanoparticles. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2007; 21:2407-14. [PMID: 17582624 DOI: 10.1002/rcm.3100] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Specific capture of phosphopeptides from protein digests is a critical step for identification of phosphoproteins by mass spectrometry. In this study, we report a novel phosphopeptide-capture approach based on the specific interaction of phosphopeptides with a stainless steel target modified with magnetic affinity nanoparticles. The modification which was carried out by loading the suspension of nanoparticles into sample wells of the target did not require any pretreatment procedure to the target and did not involve chemical binding reactions. To isolate phosphopeptides, digests were loaded into the wells of the modified target for 10 min incubation, followed by rinsing with washing buffer to remove unbound species; matrix was then added to the captured phosphopeptides prior to analysis by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOFMS). Capturing the phosphopeptides on the modified target simplified significantly analytical operations and reduced sample loss. This approach has been applied to solution digests of alpha-casein, beta-casein, and a mixture of five proteins; a number of phosphopeptides were confidently detected. Phosphopeptides from digests of 10 fmol beta-casein could be isolated and detected by MALDI-TOFMS with this method. In addition, this approach has been applied successfully to the isolation of phosphopeptides from in-gel digestive products of sub-pmol phosphoproteins after separation by sodium dodecyl sulfate/polyacrylamide gel electrophoresis (SDS-PAGE).
Collapse
Affiliation(s)
- Feng Tan
- State Key Laboratory of Proteomics-Beijing Proteome Research Center, 33 Life Park Road, Zhongguancun, Beijing 102206, China
| | | | | | | | | | | | | |
Collapse
|
36
|
Chien KY, Chang YS, Yu JS, Fan LW, Lee CW, Chi LM. Identification of a new in vivo phosphorylation site in the cytoplasmic carboxyl terminus of EBV-LMP1 by tandem mass spectrometry. Biochem Biophys Res Commun 2006; 348:47-55. [PMID: 16875669 DOI: 10.1016/j.bbrc.2006.06.188] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2006] [Accepted: 06/30/2006] [Indexed: 11/23/2022]
Abstract
Latent membrane protein 1 (LMP1), an oncogenic protein encoded by Epstein-Barr virus (EBV), has been verified to be phosphorylated in vitro by protein casein kinase 2 (CK2). In this study, we characterized the phosphorylation of the carboxyl terminus of LMP1 fused with glutathione-S-transferase (GST-LMP1c) and the FLAG-epitope-tagged LMP1 (F-LMP1) proteins expressed in HEK293T cells. Using a combination of chemical modification and tandem mass spectrometry, we detected the phosphorylation of a tryptic peptide, 191-223 amino acids, in both GST-LMP1c catalysed by CK2 and F-LMP1-expressing cell lines. Serine residues at positions 211 and 215 were determined to be the substrates of CK2 in vitro. Most importantly, the S215 phosphorylation was also detected in F-LMP1-expressing human cell lines. The phosphorylation of S215, which is located in the carboxyl-terminus activation region 1 of LMP1, provides a new insight for investigating the role and modulation of the phosphorylation of LMP1.
Collapse
Affiliation(s)
- Kun-Yi Chien
- Department of Biochemistry and Molecular Biology, Chang Gung University, Kwei-shan, Tao-Yuan, Taiwan
| | | | | | | | | | | |
Collapse
|
37
|
Moran MF, Tong J, Taylor P, Ewing RM. Emerging applications for phospho-proteomics in cancer molecular therapeutics. Biochim Biophys Acta Rev Cancer 2006; 1766:230-41. [PMID: 16889898 DOI: 10.1016/j.bbcan.2006.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2006] [Revised: 06/15/2006] [Accepted: 06/19/2006] [Indexed: 01/19/2023]
Abstract
Protein phosphorylation is a key mechanism of cell regulation in normal and cancer cells. Various new cancer drugs and drug candidates are aimed at protein kinase targets. However, selecting patients likely to respond to these treatments, even among individuals with tumors expressing validated kinase targets remains a major challenge. There exists a need for biomarkers to facilitate the monitoring of modulation of drug-targeted kinase pathways. Phospho-proteomics involves the enrichment of phosphorylated proteins from tissue, and the application of technologies such as mass spectrometry (MS) for the identification and quantification of protein phosphorylation sites. It has potential to provide pharmacodynamic readouts of disease states and cellular drug responses in tumor samples, but technical hurdles and bioinformatics challenges will need to be addressed.
Collapse
Affiliation(s)
- Michael F Moran
- Cancer Program, Hospital For Sick Children, and McLaughlin Centre for Molecular Medicine, and Banting and Best Department of Medical Research, University of Toronto, Ontario, Canada.
| | | | | | | |
Collapse
|
38
|
Abstract
Post-translational modifications define the functional and structural plasticity of proteins in archaea, prokaryotes and eukaryotes. Multi-site protein modification modulates protein activity and macromolecular interactions and is involved in a range of fundamental molecular processes. Combining state-of-the-art technologies in molecular cell biology, protein mass spectrometry and bioinformatics, it is now feasible to discover and study the structural and functional roles of distinct protein post-translational modifications.
Collapse
Affiliation(s)
- Ole N Jensen
- Protein Research Group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark.
| |
Collapse
|
39
|
Sachon E, Mohammed S, Bache N, Jensen ON. Phosphopeptide quantitation using amine-reactive isobaric tagging reagents and tandem mass spectrometry: application to proteins isolated by gel electrophoresis. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2006; 20:1127-34. [PMID: 16521170 DOI: 10.1002/rcm.2427] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Polyacrylamide gel electrophoresis is widely used for protein separation and it is frequently the final step in protein purification in biochemistry and proteomics. Using a commercially available amine-reactive isobaric tagging reagent (iTRAQ) and mass spectrometry we obtained reproducible, quantitative data from peptides derived by tryptic in-gel digestion of proteins and phosphoproteins. The protocol combines optimized reaction conditions, miniaturized peptide handling techniques and tandem mass spectrometry to quantify low- to sub-picomole amounts of (phospho)proteins that were isolated by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). Immobilized metal affinity chromatography (FeIII-IMAC) was efficient for removal of excess reagents and for enrichment of derivatized phosphopeptides prior to matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) analysis. Phosphopeptide abundance was determined by liquid chromatography/tandem mass (LC/MS/MS) using either MALDI time-of-flight/time-of-flight (TOF/TOF) MS/MS or electrospray ionization quadrupole time-of-flight (ESI-QTOF) MS/MS instruments. Chemically labeled isobaric phosphopeptides, differing only by the position of the phosphate group, were distinguished and characterized by LC/MS/MS based on their LC elution profile and distinct MS/MS spectra. We expect this quantitative mass spectrometry method to be suitable for systematic, comparative analysis of molecular variants of proteins isolated by gel electrophoresis.
Collapse
Affiliation(s)
- E Sachon
- Department of Biochemistry & Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M., Denmark
| | | | | | | |
Collapse
|