1
|
Geddis A, Mendive-Tapia L, Sujantho A, Liu E, McAughtrie S, Goodwin R, Vendrell M, Campbell CJ. Label-Free SERS Sensors for Real-Time Monitoring of Tyrosine Phosphorylation. Anal Chem 2024; 96:17978-17983. [PMID: 39472080 PMCID: PMC11561882 DOI: 10.1021/acs.analchem.4c02860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 11/13/2024]
Abstract
Dysregulation of receptor tyrosine kinases (RTKs) has been shown to correlate with cancer cell proliferation and drug resistance. Thus, monitoring the activity of RTKs at a chemical level could provide new biomedical insights and methods to assess the drug efficacy. However, direct monitoring of kinase activity is challenging and most commonly relies on in vitro techniques such as Western blotting and ELISAs. Herein, we report the development of a gold nanoparticle-based surface-enhanced Raman scattering (SERS) sensor, which allows the real-time monitoring of tyrosine phosphorylation of a reporter peptide (Axltide) by the Axl enzyme. We demonstrate that our sensor shows strong signal localization, and we are able to detect tyrosine phosphorylation of the reporter peptide through chemical phosphorylation and enzymatically with similar peak changes to those observed in the spontaneous Raman spectra. Through monitoring the SERS spectrum, we can observe changes in phosphorylation in real time.
Collapse
Affiliation(s)
- Ailsa Geddis
- EaStCHEM
School of Chemistry, The University of Edinburgh, Edinburgh EH9 3FJ, U.K.
- Centre
for Inflammation Research, The University
of Edinburgh, Edinburgh EH16 4UU, U.K.
- IRR
Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, U.K.
| | - Lorena Mendive-Tapia
- Centre
for Inflammation Research, The University
of Edinburgh, Edinburgh EH16 4UU, U.K.
- IRR
Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, U.K.
| | - Audreylia Sujantho
- EaStCHEM
School of Chemistry, The University of Edinburgh, Edinburgh EH9 3FJ, U.K.
- Centre
for Inflammation Research, The University
of Edinburgh, Edinburgh EH16 4UU, U.K.
| | - Erica Liu
- EaStCHEM
School of Chemistry, The University of Edinburgh, Edinburgh EH9 3FJ, U.K.
- Centre
for Inflammation Research, The University
of Edinburgh, Edinburgh EH16 4UU, U.K.
| | - Sarah McAughtrie
- EaStCHEM
School of Chemistry, The University of Edinburgh, Edinburgh EH9 3FJ, U.K.
| | - Richard Goodwin
- Clinical
Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB4 0WG, U.K.
| | - Marc Vendrell
- Centre
for Inflammation Research, The University
of Edinburgh, Edinburgh EH16 4UU, U.K.
- IRR
Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, U.K.
| | - Colin J. Campbell
- EaStCHEM
School of Chemistry, The University of Edinburgh, Edinburgh EH9 3FJ, U.K.
- IRR
Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, U.K.
| |
Collapse
|
2
|
Dunphy K, Dowling P, Bazou D, O’Gorman P. Current Methods of Post-Translational Modification Analysis and Their Applications in Blood Cancers. Cancers (Basel) 2021; 13:1930. [PMID: 33923680 PMCID: PMC8072572 DOI: 10.3390/cancers13081930] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/04/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
Post-translational modifications (PTMs) add a layer of complexity to the proteome through the addition of biochemical moieties to specific residues of proteins, altering their structure, function and/or localization. Mass spectrometry (MS)-based techniques are at the forefront of PTM analysis due to their ability to detect large numbers of modified proteins with a high level of sensitivity and specificity. The low stoichiometry of modified peptides means fractionation and enrichment techniques are often performed prior to MS to improve detection yields. Immuno-based techniques remain popular, with improvements in the quality of commercially available modification-specific antibodies facilitating the detection of modified proteins with high affinity. PTM-focused studies on blood cancers have provided information on altered cellular processes, including cell signaling, apoptosis and transcriptional regulation, that contribute to the malignant phenotype. Furthermore, the mechanism of action of many blood cancer therapies, such as kinase inhibitors, involves inhibiting or modulating protein modifications. Continued optimization of protocols and techniques for PTM analysis in blood cancer will undoubtedly lead to novel insights into mechanisms of malignant transformation, proliferation, and survival, in addition to the identification of novel biomarkers and therapeutic targets. This review discusses techniques used for PTM analysis and their applications in blood cancer research.
Collapse
Affiliation(s)
- Katie Dunphy
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Paul Dowling
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Despina Bazou
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland;
| | - Peter O’Gorman
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland;
| |
Collapse
|
3
|
Lorentzian A, Uzozie A, Lange PF. Origins and clinical relevance of proteoforms in pediatric malignancies. Expert Rev Proteomics 2019; 16:185-200. [PMID: 30700156 DOI: 10.1080/14789450.2019.1575206] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Cancer changes the proteome in complex ways that reach well beyond simple changes in protein abundance. Genomic and transcriptional variations and post-translational protein modification create functional variants of a protein, known as proteoforms. Childhood cancers have fewer genomic alterations but show equally dramatic phenotypic changes as malignant cells in adults. Therefore, unraveling the complexities of the proteome is even more important in pediatric malignancies. Areas covered: In this review, the biological origins of proteoforms and technological advancements in the study of proteoforms are discussed. Particular emphasis is given to their implication in childhood malignancies and the critical role of cancer-specific proteoforms for the next generation of cancer therapies and diagnostics. Expert opinion: Recent advancements in technology have led to a better understanding of the underlying mechanisms of tumorigenesis. This has been critical for the development of more effective and less harmful treatments that are based on direct targeting of altered proteins and deregulated pathways. As proteome coverage and the ability to detect complex proteoforms increase, the most need for change is in data compilation and database availability to mediate high-level data analysis and allow for better functional annotation of proteoforms.
Collapse
Affiliation(s)
- Amanda Lorentzian
- a Department of Cell and Developmental Biology , University of British Columbia , Vancouver , BC , Canada.,b Michael Cuccione Childhood Cancer Research Program , BC Children's Hospital Research Institute , Vancouver , BC , Canada
| | - Anuli Uzozie
- b Michael Cuccione Childhood Cancer Research Program , BC Children's Hospital Research Institute , Vancouver , BC , Canada.,c Department of Pathology and Laboratory Medicine , University of British Columbia , Vancouver , BC , Canada
| | - Philipp F Lange
- a Department of Cell and Developmental Biology , University of British Columbia , Vancouver , BC , Canada.,b Michael Cuccione Childhood Cancer Research Program , BC Children's Hospital Research Institute , Vancouver , BC , Canada.,c Department of Pathology and Laboratory Medicine , University of British Columbia , Vancouver , BC , Canada
| |
Collapse
|
4
|
Yang S, Guo Y, Zhang W, Zhang J, Zhang Y, Xu P. Effect of FGF-21 on implant bone defects through hepatocyte growth factor (HGF)-mediated PI3K/AKT signaling pathway. Biomed Pharmacother 2018; 109:1259-1267. [PMID: 30551376 DOI: 10.1016/j.biopha.2018.10.150] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 10/22/2018] [Accepted: 10/25/2018] [Indexed: 11/28/2022] Open
Abstract
Implant bone defects are the most common phenomenon in the processes of bone transplantation. Evidences have identified that fibroblast growth factor-21 (FGF-21) encourages osteogenesis for patients with implant bone defects. The purpose of this study was to investigate the role of FGF-21 and its potential mechanism in bone mesenchymal stem cells (BMSCs). RT-PCR, Western blotting, flow cytometry, immunofluorescence and immunohistochemistry assays were performed to analyze the role of FGF-21 and intracellular signaling pathways involved in BMSCs. It was shown that FGF-21 increased viability of BMSCs. Treatment with FGF-21 decreased the apoptosis of BMSCs by decreasing pro-apoptosis protein Caspase-3. Results indicated that FGF-21 (2 mg/kg) treatment up-regulated HGF, PI3K and AKT expression in BMSCs. In addition, the protective effects of FGF-21 on BMSCs were canceled by PI3K/AKT inhibitor in BMSCs. Results found that knockdown of HGF abolished FGF-21-decreased PI3K/AKT signal pathway. Furthermore, results demonstrated that FGF-21 presented beneficial effects for implant bone defects in rat model. In conclusion, these results indicate that FGF-21 can improve implant bone defects through HGF-mediated PI3K/AKT signaling pathway in BMSCs.
Collapse
Affiliation(s)
- Shimao Yang
- Department of Oral and Maxillofacial Surgery, Jinan Stomatology Hospital, Jinan City, Shandong Province, China
| | - Yanwei Guo
- Department of Oral and Maxillofacial Surgery, Jining Stomatology Hospital, Jining City, Shandong Province, China
| | - Wenmei Zhang
- Department of Oral and Maxillofacial Surgery, Jinan Stomatology Hospital, Jinan City, Shandong Province, China
| | - Jin Zhang
- Department of Oral and Maxillofacial Surgery, Jinan Stomatology Hospital, Jinan City, Shandong Province, China
| | - Yujie Zhang
- Department of Oral and Maxillofacial Surgery, Jinan Stomatology Hospital, Jinan City, Shandong Province, China
| | - Peng Xu
- Department of Dental Implant, Stomatological Hospital of Chongqing Medical University, Chongqing, 400015, China.
| |
Collapse
|
5
|
Gai J, Gao Z, Song L, Xu Y, Liu W, Zhao C. Contrast-enhanced computed tomography combined with Chitosan-Fe 3O 4 nanoparticles targeting fibroblast growth factor receptor and vascular endothelial growth factor receptor in the screening of early esophageal cancer. Exp Ther Med 2018; 15:5344-5352. [PMID: 29805549 PMCID: PMC5958695 DOI: 10.3892/etm.2018.6087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 04/28/2017] [Indexed: 12/14/2022] Open
Abstract
Esophageal cancer is a malignant tumor with a relatively high invasiveness, metastatic potential and worldwide incidence among human cancers. The majority of patients with esophageal cancer are diagnosed in a late tumor stage due to a lack of advanced and sensitive protocols for the diagnosis of patients with early-stage esophageal cancer. In the current study, contrast-enhanced computerized tomography (CECT) combined with Chitosan-Fe3O4 nanoparticles targeting fibroblast growth factor receptor (FGFR) and vascular endothelial growth factor receptor (VEGFR; CECT-CNFV) were used to diagnose patients with suspected esophageal cancer. A Chitosan-Fe3O4-parceled bispecific antibody targeting FGFR and VEGFR was produced and its affinity to esophageal cancer cells was determined both in vitro and in vivo. A total of 320 patients with suspected esophageal cancer were voluntarily recruited to evaluate the efficacy of CECT-CNFV in the diagnosis of early-stage esophageal cancer. All participants were subjected to CT and CECT-CNFV to detect whether tumors were present in the esophageal area. A Chitosan-Fe3O4 nanoparticles contrast agent was orally administered at 20 min prior to CT and CECT-CNFV. The results demonstrated that CECT-CNFV improved diagnostic sensitivity and provided a novel protocol for the diagnosis of tumors in patients with suspected gastric cancer at an early-stage. Furthermore, the resolution ratio of images was enhanced by CECT-CNFV, which enabled the visualization of tiny tumor nodules in esophageal tissue. Clinical data demonstrated that CECT-CNFV diagnosed 200 patients with suspected early-stage esophageal cancer and 120 patients as tumor free. In addition, CECT-CNFV exhibited higher signal enhancement of tumor nodules than CT, suggesting a higher accuracy and accumulation of nanoparticle contrast agent within the tumor nodules of esophageal tissue. Notably, the survival rate of patients with esophageal cancer diagnosed at an early-stage by CECT-CNFV was higher than the mean five-year survival rate (P<0.01). In conclusion, CECT-CNFV enhanced the sensitivity and accuracy of CT in the diagnosis of early-stage esophageal cancer. Thus, CECT-CNFV may improve the accuracy of CT in the diagnosis of mural enhancement in patients with esophageal cancer.
Collapse
Affiliation(s)
- Juanjuan Gai
- Department of Radiology, Dongying People's Hospital, Dongying, Shandong 257091, P.R. China
| | - Zhenli Gao
- Department of Radiology, Dongying People's Hospital, Dongying, Shandong 257091, P.R. China
| | - Liqiang Song
- Department of Oncology, Dongying People's Hospital, Dongying, Shandong 257091, P.R. China
| | - Yongyun Xu
- Department of Computed Tomography, Dongying People's Hospital, Dongying, Shandong 257091, P.R. China
| | - Weixin Liu
- Department of Oncology, Dongying People's Hospital, Dongying, Shandong 257091, P.R. China
| | - Chuanxin Zhao
- Department of Joint Surgery, Dongying People's Hospital, Dongying, Shandong 257091, P.R. China
| |
Collapse
|
6
|
Guo YE, Riley KJ, Iwasaki A, Steitz JA. Alternative capture of noncoding RNAs or protein-coding genes by herpesviruses to alter host T cell function. Mol Cell 2014; 54:67-79. [PMID: 24725595 DOI: 10.1016/j.molcel.2014.03.025] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 02/10/2014] [Accepted: 03/01/2014] [Indexed: 11/25/2022]
Abstract
In marmoset T cells transformed by Herpesvirus saimiri (HVS), a viral U-rich noncoding (nc) RNA, HSUR 1, specifically mediates degradation of host microRNA-27 (miR-27). High-throughput sequencing of RNA after crosslinking immunoprecipitation (HITS-CLIP) identified mRNAs targeted by miR-27 as enriched in the T cell receptor (TCR) signaling pathway, including GRB2. Accordingly, transfection of miR-27 into human T cells attenuates TCR-induced activation of mitogen-activated protein kinases (MAPKs) and induction of CD69. MiR-27 also robustly regulates SEMA7A and IFN-γ, key modulators and effectors of T cell function. Knockdown or ectopic expression of HSUR 1 alters levels of these proteins in virally transformed cells. Two other T-lymphotropic γ-herpesviruses, AlHV-1 and OvHV-2, do not produce a noncoding RNA to downregulate miR-27 but instead encode homologs of miR-27 target genes. Thus, oncogenic γ-herpesviruses have evolved diverse strategies to converge on common targets in host T cells.
Collapse
Affiliation(s)
- Yang Eric Guo
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Kasandra J Riley
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06536, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Joan A Steitz
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT 06536, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06536, USA.
| |
Collapse
|
7
|
Okur MN, Russo A, O'Bryan JP. Receptor tyrosine kinase ubiquitylation involves the dynamic regulation of Cbl-Spry2 by intersectin 1 and the Shp2 tyrosine phosphatase. Mol Cell Biol 2014; 34:271-9. [PMID: 24216759 PMCID: PMC3911288 DOI: 10.1128/mcb.00850-13] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 07/30/2013] [Accepted: 10/31/2013] [Indexed: 11/20/2022] Open
Abstract
Ubiquitylation of receptor tyrosine kinases (RTKs) regulates their trafficking and lysosomal degradation. The multidomain scaffolding protein intersectin 1 (ITSN1) is an important regulator of this process. ITSN1 stimulates ubiquitylation of the epidermal growth factor receptor (EGFR) through enhancing the activity of the Cbl E3 ubiquitin ligase. However, the precise mechanism through which ITSN1 enhances Cbl activity is unclear. Here, we demonstrate that ITSN1 interacts with and recruits the Shp2 tyrosine phosphatase to Spry2 to enhance its dephosphorylation, thereby disrupting the inhibitory effect of Spry2 on Cbl and enhancing EGFR ubiquitylation. In contrast, expression of a catalytically inactive Shp2 mutant reversed the effect of ITSN1 on Spry2 dephosphorylation and decreased Cbl-mediated EGFR ubiquitylation. In addition, disruption of ITSN1 binding to Spry2 through point mutation of the Pro-rich ITSN1 binding site in Spry2 resulted in decreased Shp2-Spry2 interaction and enhanced Spry2 tyrosine phosphorylation. This study demonstrates that ITSN1 enhances Cbl activity, in part, by modulating the interaction of Cbl with Spry2 through recruitment of Shp2 phosphatase to the Cbl-Spry2 complex. These findings reveal a new level of complexity in the regulation of RTKs by Cbl through ITSN1 binding with Shp2 and Spry2.
Collapse
Affiliation(s)
- Mustafa Nazir Okur
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Angela Russo
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - John P. O'Bryan
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, Illinois, USA
- Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|