101
|
Protein Clusters in Phosphotyrosine Signal Transduction. J Mol Biol 2018; 430:4547-4556. [PMID: 29870724 DOI: 10.1016/j.jmb.2018.05.040] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/08/2018] [Accepted: 05/28/2018] [Indexed: 11/20/2022]
Abstract
Signal transduction systems based on tyrosine phosphorylation are central to cell-cell communication in multicellular organisms. Typically, in such a system, the signal is initiated by activating tyrosine kinases associated with transmembrane receptors, which induces tyrosine phosphorylation of the receptor and/or associated proteins. The phosphorylated tyrosines then serve as docking sites for the binding of various downstream effector proteins. It has long been observed that the cooperative association of the receptors and effectors produces higher-order protein assemblies (clusters) following signal activation in virtually all phosphotyrosine signal transduction systems. However, mechanistic studies on how such clustering processes affect signal transduction outcomes have only emerged recently. Here we review current progress in decoding the biophysical consequences of clustering on the behavior of the system, and how clustering affects how these receptors process information.
Collapse
|
102
|
Synthetic cytokine receptors transmit biological signals using artificial ligands. Nat Commun 2018; 9:2034. [PMID: 29789554 PMCID: PMC5964073 DOI: 10.1038/s41467-018-04454-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/23/2018] [Indexed: 11/25/2022] Open
Abstract
Cytokine-induced signal transduction is executed by natural biological switches, which among many others control immune-related processes. Here, we show that synthetic cytokine receptors (SyCyRs) can induce cytokine signaling using non-physiological ligands. High-affinity GFP- and mCherry-nanobodies were fused to transmembrane and intracellular domains of the IL-6/IL-11 and IL-23 cytokine receptors gp130 and IL-12Rβ1/IL-23R, respectively. Homo- and heterodimeric GFP:mCherry fusion proteins as synthetic cytokine-like ligands were able to induce canonical signaling in vitro and in vivo. Using SyCyR ligands, we show that IL-23 receptor homodimerization results in its activation and IL-23-like signal transduction. Moreover, trimeric receptor assembly induces trans-phosphorylation among cytokine receptors with associated Janus kinases. The SyCyR technology allows biochemical analyses of transmembrane receptor signaling in vitro and in vivo, cell-specific activation through SyCyR ligands using transgenic animals and possible therapeutic regimes involving non-physiological targets during immunotherapy. Cytokine-induced signaling acts as an ON/OFF switch dependent on the presence of ligands. Here the authors construct synthetic cytokine receptors responsive to synthetic ligands able to activate canonical signaling pathways.
Collapse
|
103
|
Abstract
Engineering synthetic gene regulatory circuits proceeds through iterative cycles of design, building, and testing. Initial circuit designs must rely on often-incomplete models of regulation established by fields of reductive inquiry—biochemistry and molecular and systems biology. As differences in designed and experimentally observed circuit behavior are inevitably encountered, investigated, and resolved, each turn of the engineering cycle can force a resynthesis in understanding of natural network function. Here, we outline research that uses the process of gene circuit engineering to advance biological discovery. Synthetic gene circuit engineering research has not only refined our understanding of cellular regulation but furnished biologists with a toolkit that can be directed at natural systems to exact precision manipulation of network structure. As we discuss, using circuit engineering to predictively reorganize, rewire, and reconstruct cellular regulation serves as the ultimate means of testing and understanding how cellular phenotype emerges from systems-level network function.
Collapse
Affiliation(s)
- Caleb J. Bashor
- Institute for Medical Engineering and Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;,
| | - James J. Collins
- Institute for Medical Engineering and Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;,
- Harvard–MIT Program in Health Sciences and Technology, Cambridge, Massachusetts 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, USA
| |
Collapse
|
104
|
Zebrafish VCAP1X2 regulates cardiac contractility and proliferation of cardiomyocytes and epicardial cells. Sci Rep 2018; 8:7856. [PMID: 29777134 PMCID: PMC5959901 DOI: 10.1038/s41598-018-26110-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/01/2018] [Indexed: 01/08/2023] Open
Abstract
Sarcomeric signaling complexes are important to sustain proper sarcomere structure and function, however, the mechanisms underlying these processes are not fully elucidated. In a gene trap experiment, we found that vascular cell adhesion protein 1 isoform X2 (VCAP1X2) mutant embryos displayed a dilated cardiomyopathy phenotype, including reduced cardiac contractility, enlarged ventricular chamber and thinned ventricular compact layer. Cardiomyocyte and epicardial cell proliferation was decreased in the mutant heart ventricle, as was the expression of pAKT and pERK. Contractile dysfunction in the mutant was caused by sarcomeric disorganization, including sparse myofilament, blurred Z-disc, and decreased gene expression for sarcomere modulators (smyd1b, mypn and fhl2a), sarcomeric proteins (myh6, myh7, vmhcl and tnnt2a) and calcium regulators (ryr2b and slc8a1a). Treatment of PI3K activator restored Z-disc alignment while injection of smyd1b mRNA restored Z-disc alignment, contractile function and cardiomyocyte proliferation in ventricles of VCAP1X2 mutant embryos. Furthermore, injection of VCAP1X2 variant mRNA rescued all phenotypes, so long as two cytosolic tyrosines were left intact. Our results reveal two tyrosine residues located in the VCAP1X2 cytoplasmic domain are essential to regulate cardiac contractility and the proliferation of ventricular cardiomyocytes and epicardial cells through modulating pAKT and pERK expression levels.
Collapse
|
105
|
Xu YZ, Thuraisingam T, Kanagaratham C, Tao S, Radzioch D. c-Src kinase is involved in the tyrosine phosphorylation and activity of SLC11A1 in differentiating macrophages. PLoS One 2018; 13:e0196230. [PMID: 29723216 PMCID: PMC5933793 DOI: 10.1371/journal.pone.0196230] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 04/09/2018] [Indexed: 11/18/2022] Open
Abstract
Studies have demonstrated that the solute carrier family 11 member 1 (SLC11A1) is heavily glycosylated and phosphorylated in macrophages. However, the mechanisms of SLC11A1 phosphorylation, and the effects of phosphorylation on SLC11A1 activity remain largely unknown. Here, the tyrosine phosphorylation of SLC11A1 is observed in SLC11A1-expressing U937 cells when differentiated into macrophages by phorbol myristate acetate (PMA). The phosphorylation of SLC11A1 is almost completely blocked by treatment with PP2, a selective inhibitor of Src family kinases. Furthermore, we found that SLC11A1 is a direct substrate for active c-Src kinase and siRNA-mediated knockdown of cellular Src (c-Src) expression results in a significant decrease in tyrosine phosphorylation. We found that PMA induces the interaction of SLC11A1 with c-Src kinase. We demonstrated that SLC11A1 is phosphorylated by Src family kinases at tyrosine 15 and this type of phosphorylation is required for SLC11A1-mediated modulation of NF-κB activation and nitric oxide (NO) production induced by LPS. Our results demonstrate important roles for c-Src tyrosine kinase in phosphorylation and activation of SLC11A1 in macrophages.
Collapse
Affiliation(s)
- Yong Zhong Xu
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Thusanth Thuraisingam
- Division of Dermatology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Cynthia Kanagaratham
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Shao Tao
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Danuta Radzioch
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
- * E-mail:
| |
Collapse
|
106
|
Evolutionary convergence and divergence in archaeal chromosomal proteins and Chromo-like domains from bacteria and eukaryotes. Sci Rep 2018; 8:6196. [PMID: 29670199 PMCID: PMC5906684 DOI: 10.1038/s41598-018-24467-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/04/2018] [Indexed: 11/08/2022] Open
Abstract
SH3-fold-β-barrel domains of the chromo-like superfamily recognize epigenetic marks in eukaryotic proteins. Their provenance has been placed either in archaea, based on apparent structural similarity to chromatin-compacting Sul7d and Cren7 proteins, or in bacteria based on the presence of sequence homologs. Using sequence and structural evidence we establish that the archaeal Cren7/Sul7 proteins emerged from a zinc ribbon (ZnR) ancestor. Further, we show that the ancestral eukaryotic chromo-like domains evolved from bacterial versions, likely acquired from early endosymbioses, which already possessed an aromatic cage for recognition of modified amino-groups. These bacterial versions are part of a radiation of secreted SH3-fold domains, which spawned both chromo-like domains and classical SH3 domains in the context of peptide-recognition in the peptidoglycan or the extracellular matrix. This establishes that Cren7/Sul7 converged to a “SH3”-like state from a ZnR precursor via the loss of metal-chelation and acquisition of stronger hydrophobic interactions; it is unlikely to have participated in the evolution of the chromo-like domains. We show that archaea possess several Cren7/Sul7-related proteins with intact Zn-chelating ligands, which we predict to play previously unstudied roles in chromosome segregation during cell-division comparable to the PRC barrel and CdvA domain proteins.
Collapse
|
107
|
Kieken F, Loth K, van Nuland N, Tompa P, Lenaerts T. Chemical shift assignments of the partially deuterated Fyn SH2-SH3 domain. BIOMOLECULAR NMR ASSIGNMENTS 2018; 12:117-122. [PMID: 29224116 DOI: 10.1007/s12104-017-9792-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 11/28/2017] [Indexed: 06/07/2023]
Abstract
Src Homology 2 and 3 (SH2 and SH3) are two key protein interaction modules involved in regulating the activity of many proteins such as tyrosine kinases and phosphatases by respective recognition of phosphotyrosine and proline-rich regions. In the Src family kinases, the inactive state of the protein is the direct result of the interaction of the SH2 and the SH3 domain with intra-molecular regions, leading to a closed structure incompetent with substrate modification. Here, we report the 1H, 15N and 13C backbone- and side-chain chemical shift assignments of the partially deuterated Fyn SH3-SH2 domain and structural differences between tandem and single domains. The BMRB accession number is 27165.
Collapse
Affiliation(s)
- Fabien Kieken
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussel, Belgium
- Center for Structural Biology, VIB, Pleinlaan 2, 1050, Brussel, Belgium
- AI-lab, Vakgroep Computerwetenschappen, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
- Interuniversity Institute of Bioinformatics in Brussels (IB2), ULB-VUB, La Plaine Campus, Boulevard du Triomphe, CP 263, 1050, Brussels, Belgium
| | - Karine Loth
- Centre de Biophysique Moléculaire, Centre National de la Recherche Scientifique (CNRS) UPR 4301, Université d'Orléans, rue Charles Sadron, 45071, Orléans Cedex 2, France
- Collegium Sciences et Techniques, Université d'Orléans, rue de Chartres, 45100, Orléans, France
| | - Nico van Nuland
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussel, Belgium
- Center for Structural Biology, VIB, Pleinlaan 2, 1050, Brussel, Belgium
| | - Peter Tompa
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussel, Belgium
- Center for Structural Biology, VIB, Pleinlaan 2, 1050, Brussel, Belgium
| | - Tom Lenaerts
- AI-lab, Vakgroep Computerwetenschappen, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium.
- Interuniversity Institute of Bioinformatics in Brussels (IB2), ULB-VUB, La Plaine Campus, Boulevard du Triomphe, CP 263, 1050, Brussels, Belgium.
- MLG, Départment d'Informatique, Université Libre de Bruxelles, Boulevard du Triomphe, CP 212, 1050, Brussels, Belgium.
| |
Collapse
|
108
|
Downregulated SASH1 expression indicates poor clinical prognosis in gastric cancer. Hum Pathol 2018; 74:83-91. [DOI: 10.1016/j.humpath.2018.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 12/20/2017] [Accepted: 01/02/2018] [Indexed: 02/07/2023]
|
109
|
Characterization of two splice variants of EGFR and their effects on the growth of the razor clam. AQUACULTURE AND FISHERIES 2018. [DOI: 10.1016/j.aaf.2018.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
110
|
Brown T, Brown N, Stollar EJ. Most yeast SH3 domains bind peptide targets with high intrinsic specificity. PLoS One 2018; 13:e0193128. [PMID: 29470497 PMCID: PMC5823434 DOI: 10.1371/journal.pone.0193128] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/04/2018] [Indexed: 01/07/2023] Open
Abstract
A need exists to develop bioinformatics for predicting differences in protein function, especially for members of a domain family who share a common fold, yet are found in a diverse array of proteins. Many domain families have been conserved over large evolutionary spans and representative genomic data during these periods are now available. This allows a simple method for grouping domain sequences to reveal common and unique/specific binding residues. As such, we hypothesize that sequence alignment analysis of the yeast SH3 domain family across ancestral species in the fungal kingdom can determine whether each member encodes specific information to bind unique peptide targets. With this approach, we identify important specific residues for a given domain as those that show little conservation within an alignment of yeast domain family members (paralogs) but are conserved in an alignment of its direct relatives (orthologs). We find most of the yeast SH3 domain family members have maintained unique amino acid conservation patterns that suggest they bind peptide targets with high intrinsic specificity through varying degrees of non-canonical recognition. For a minority of domains, we predict a less diverse binding surface, likely requiring additional factors to bind targets specifically. We observe that our predictions are consistent with high throughput binding data, which suggests our approach can probe intrinsic binding specificity in any other interaction domain family that is maintained during evolution.
Collapse
Affiliation(s)
- Tom Brown
- Math and Computer Science Department, Eastern New Mexico University, Portales, NM, United States of America
| | - Nick Brown
- Portales High School, Portales, NM, United States of America
| | - Elliott J. Stollar
- Physical Sciences Department, Eastern New Mexico University, Portales, NM, United States of America
- * E-mail:
| |
Collapse
|
111
|
Abstract
Src Homology 2 (SH2) domains are protein interaction modules that recognize and bind tyrosine phosphorylated ligands. Their ability to distinguish binding to over thousands of potential phosphotyrosine (pTyr) ligands within the cell is critical for the fidelity of receptor tyrosine kinase (RTK) signaling. Within humans there are over a hundred SH2 domains with more than several thousand potential ligands across many cell types and cell states. Therefore, defining the specificity of individual SH2 domains is critical for predicting and identifying their physiological ligands. Here, in this chapter, I describe the broad use of SPOT peptide arrays for examining SH2 domain specificity. An orientated peptide array library (OPAL) approach can uncover both favorable and non-favorable residues, thus providing an in-depth analysis to SH2 specificity. Moreover, I discuss the application of SPOT arrays for paneling SH2 ligand binding with physiological peptides.
Collapse
|
112
|
Structural and functional modeling of viral protein 5 of Infectious Bursal Disease Virus. Virus Res 2018; 247:55-60. [PMID: 29427596 DOI: 10.1016/j.virusres.2018.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/28/2018] [Accepted: 01/31/2018] [Indexed: 11/22/2022]
Abstract
Infectious Bursal Disease (IBD) is an acute, highly contagious and immunosuppressive disease of young chicken. The causative virus (IBDV) is a bi-segmented, double-stranded RNA virus. The virus encodes five major proteins, viral protein (VP) 1-5. VPs 1-3 have been characterized crystallographically. Albeit a rise in the number of studies reporting successful heterologous expression of VP5 in recent times, challenging the notion that rapid death of host cells overexpressing VP5 disallows obtaining sufficiently pure preparations of the protein for crystallographic studies, the structure of VP5 remains unknown and its function controversial. Our study describes the first 3D model of IBD VP5 obtained through an elaborate computational workflow. Based on the results of the study, IBD VP5 can be predicted to be a structural analog of the leucine-rich repeat (LRR) family of proteins. Functional implications arising from structural similarity of VP5 with host Toll-like receptor (Tlr) 3 also satisfy the previously reported opposing roles of the protein in first abolishing and later inducing host-cell apoptosis.
Collapse
|
113
|
Structure function relations in PDZ-domain-containing proteins: Implications for protein networks in cellular signalling. J Biosci 2017. [DOI: 10.1007/s12038-017-9727-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
114
|
The regulatory role of protein phosphorylation in human gammaherpesvirus associated cancers. Virol Sin 2017; 32:357-368. [PMID: 29116588 PMCID: PMC6704201 DOI: 10.1007/s12250-017-4081-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022] Open
Abstract
Activation of specific sets of protein kinases by intracellular signal molecules
has become more and more apparent in the past decade. Phosphorylation, one of key
posttranslational modification events, is activated by kinase or regulatory protein
and is vital for controlling many physiological functions of eukaryotic cells such
as cell proliferation, differentiation, malignant transformation, and signal
transduction mediated by external stimuli. Moreovers, the reversible modification of
phosphorylation and dephosphorylation can result in different features of the target
substrate molecules including DNA binding, protein-protein interaction, subcellular
location and enzymatic activity, and is often hijacked by viral infection.
Epstein-Barr virus (EBV) and Kaposi’s sarcomaassociated herpesvirus (KSHV), two
human oncogenic gamma-herpesviruses, are shown to tightly associate with many
malignancies. In this review, we summarize the recent progresses on understanding of
molecular properties and regulatory modes of cellular and viral proteins
phosphorylation influenced by these two tumor viruses, and highlight the potential
therapeutic targets and strategies against their related cancers. ![]()
Collapse
|
115
|
Bhakta HK, Paudel P, Fujii H, Sato A, Park CH, Yokozawa T, Jung HA, Choi JS. Oligonol promotes glucose uptake by modulating the insulin signaling pathway in insulin-resistant HepG2 cells via inhibiting protein tyrosine phosphatase 1B. Arch Pharm Res 2017; 40:1314-1327. [PMID: 29027136 DOI: 10.1007/s12272-017-0970-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 09/19/2017] [Indexed: 11/26/2022]
Abstract
Insulin resistance and protein tyrosine phosphatase 1B (PTP1B) overexpression are strongly associated with type 2 diabetes mellitus (T2DM), which is characterized by defects in insulin signaling and glucose intolerance. In a previous study, we demonstrated oligonol inhibits PTP1B and α-glucosidase related to T2DM. In this study, we examined the molecular mechanisms underlying the anti-diabetic effects of oligonol in insulin-resistant HepG2 cells. Glucose uptake was assessed using a fluorescent glucose tracer, 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-deoxyglucose, and the signaling pathway was investigated by western blotting. Oligonol significantly increased insulin-provoked glucose uptake and decreased PTP1B expression, followed by modulation of ERK phosphorylation. In addition, oligonol activated insulin receptor substrate 1 by reducing phosphorylation at serine 307 and increasing that at tyrosine 895, and enhanced the phosphorylations of Akt and phosphatidylinositol 3-kinase. Interestingly, it also reduced the expression of two key enzymes of gluconeogenesis (glucose 6-phosphatase and phosphoenolpyruvate carboxykinase), attenuated oxidative stress by scavenging/inhibiting peroxynitrite, and reactive oxygen species (ROS) generation, and augmented the expression of nuclear factor kappa B. These findings suggest oligonol improved the insulin sensitivity of insulin-resistant HepG2 cells by attenuating the insulin signaling blockade and modulating glucose uptake and production. Furthermore, oligonol attenuated ROS-related inflammation and prevented oxidative damage in our in vitro model of type 2 diabetes. These result indicate oligonol has promising potential as a treatment for T2DM.
Collapse
Affiliation(s)
- Himanshu Kumar Bhakta
- Department of Food and Life Science, Pukyong National University, Busan, 608-737, Republic of Korea
| | - Pradeep Paudel
- Department of Food and Life Science, Pukyong National University, Busan, 608-737, Republic of Korea
| | - Hajime Fujii
- Amino Up Chemical Company Ltd., Sapporo, 004-0839, Japan
| | - Atsuya Sato
- Amino Up Chemical Company Ltd., Sapporo, 004-0839, Japan
| | - Chan Hum Park
- Department of Medicinal Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration, Eumseong, 369-873, Republic of Korea
| | - Takako Yokozawa
- Graduate School of Science and Engineering for Research, University of Toyama, Toyama, 930-8555, Japan
| | - Hyun Ah Jung
- Department of Food Science and Human Nutrition, Chonbuk National University, Jeonju, 561-756, Republic of Korea.
| | - Jae Sue Choi
- Department of Food and Life Science, Pukyong National University, Busan, 608-737, Republic of Korea.
| |
Collapse
|
116
|
Identification and characterization of tyrosine kinases in anole lizard indicate the conserved tyrosine kinase repertoire in vertebrates. Mol Genet Genomics 2017; 292:1405-1418. [PMID: 28819830 DOI: 10.1007/s00438-017-1356-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/08/2017] [Indexed: 10/19/2022]
Abstract
The tyrosine kinases (TKs) play principal roles in regulation of multicellular aspects of the organism and are implicated in many cancer types and congenital disorders. The anole lizard has recently been introduced as a model organism for laboratory-based studies of organismal function and field studies of ecology and evolution. However, the TK family of anole lizard has not been systematically identified and characterized yet. In this study, we identified 82 TK-encoding genes in the anole lizard genome and classified them into 28 subfamilies through phylogenetic analysis, with no member from ROS and STYK1 subfamilies identified. Although TK domain sequences and domain organization in each subfamily were conserved, the total number of TKs in different species was much variable. In addition, extensive evolutionary analysis in metazoans indicated that TK repertoire in vertebrates tends to be remarkably stable. Phylogenetic analysis of Eph subfamily indicated that the divergence of EphA and EphB occurred prior to the whole genome duplication (WGD) but after the split of Urochordates and vertebrates. Moreover, the expression pattern analysis of lizard TK genes among 9 different tissues showed that 14 TK genes exhibited tissue-specific expression and 6 TK genes were widely expressed. Comparative analysis of TK expression suggested that the tissue specifically expressed genes showed different expression pattern but the widely expressed genes showed similar pattern between anole lizard and human. These results may provide insights into the evolutionary diversification of animal TK genes and would aid future studies on TK protein regulation of key growth and developmental processes.
Collapse
|
117
|
Papanastasiou AD, Sirinian C, Plakoula E, Zolota V, Zarkadis IK, Kalofonos HP. RANK and EGFR in invasive breast carcinoma. Cancer Genet 2017; 216-217:61-66. [PMID: 29025596 DOI: 10.1016/j.cancergen.2017.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 01/16/2023]
Abstract
Breast cancer is the most common malignancy, affecting one in eight women in North America and Europe. The human epidermal growth factor receptor (EGFR) protein comprises a major determinant of normal development but also cancer. RANK receptor (Receptor Activator of Nuclear factor-κB) is a tumor necrosis superfamily member and a binding partner for RANKL, which was recently implicated in breast cancer initiation, progression and metastasis. Here we provide preliminary evidence of a possible interplay between RANK and EGFR signaling in breast cancer. TCGA (cancergenome.nih.gov) publicly available data for EGFR and TNFRSF11A (RANK) genes from breast cancer patients and breast cancer cell lines were retrieved and analyzed. RANK mRNA showed a statistically significant positive correlation (p <0.001) with the mRNA and protein expression of EGFR, but not with ERBB2/3/4. Further analyses of survival data of a group of breast cancer patients (n = 248) from TCGA, revealed an EGFRhi/RANKhi subpopulation that showed a statistically significant (p = 0.001) reduced overall survival when compared to EGFRlow/RANKlow group of patients. Finally, EGFR and RANK combinatorial in vitro analyses revealed a significant upregulation of AKT and ERK signaling after EGF stimulation in cell lines and also an increase of breast cancer cell invasiveness.
Collapse
Affiliation(s)
- Anastasios D Papanastasiou
- Clinical and Molecular Oncology Laboratory, Division of Oncology, School of Medicine, University of Patras, 26504, Greece.
| | - Chaido Sirinian
- Clinical and Molecular Oncology Laboratory, Division of Oncology, School of Medicine, University of Patras, 26504, Greece
| | - Eva Plakoula
- Clinical and Molecular Oncology Laboratory, Division of Oncology, School of Medicine, University of Patras, 26504, Greece; Department of Biology, School of Medicine, University of Patras, 26504, Greece
| | - Vassiliki Zolota
- Department of Pathology, School of Medicine, University of Patras, 26504, Greece
| | - Ioannis K Zarkadis
- Department of Biology, School of Medicine, University of Patras, 26504, Greece
| | - Haralabos P Kalofonos
- Clinical and Molecular Oncology Laboratory, Division of Oncology, School of Medicine, University of Patras, 26504, Greece
| |
Collapse
|
118
|
van der Meulen T, Swarts S, Fischer W, van der Geer P. Identification of STS-1 as a novel ShcA-binding protein. Biochem Biophys Res Commun 2017; 490:1334-1339. [PMID: 28690151 DOI: 10.1016/j.bbrc.2017.07.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/05/2017] [Indexed: 11/19/2022]
Abstract
ShcA is a cytoplasmic signaling protein that supports signal transduction by receptor protein-tyrosine kinases by providing auxiliary tyrosine phosphorylation sites that engage additional signaling proteins. The principal binding partner for tyrosine phosphorylation sites on ShcA is Grb2. In the current study, we have used phosphotyrosine-containing peptides to isolate and identify STS-1 as a novel ShcA-binding protein. Our results further show that the interaction between STS-1 and ShcA is regulated in response to EGF receptor activation.
Collapse
Affiliation(s)
- Talitha van der Meulen
- Department of Chemistry and Biochemistry, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182-1030, USA
| | - Spencer Swarts
- Department of Chemistry and Biochemistry, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182-1030, USA
| | - Wolfgang Fischer
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute, 10010 North Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Peter van der Geer
- Department of Chemistry and Biochemistry, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182-1030, USA.
| |
Collapse
|
119
|
Almawi WY, Hess DA, Rieder MJ. Multiplicity of Glucocorticoid Action in Inhibiting Allograft Rejection. Cell Transplant 2017; 7:511-23. [PMID: 9853580 DOI: 10.1177/096368979800700602] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Glucocorticoids (GCs) are used as immunosuppressive and antiinflammatory agents in organ transplantation and in treating autoimmune diseases and inflammatory disorders. GCs were shown to exert their antiproliferative effects directly through blockade of certain elements of an early membrane-associated signal transduction pathway, modulation of the expression of select adhesion molecules, and by suppression of cytokine synthesis and action. GCs may act indirectly by inducing lipocortin synthesis, which in turn, inhibits arachidonic acid release from membrane-bound stores, and also by inducing transforming growth factor (TGF)-β expression that subsequently blocks cytokine synthesis and T cell activation. Furthermore, by preferentially inhibiting the production of Th1 cytokines, GCs may enhance Th2 cell activity and, hence, precipitate a long-lasting state of tolerance through a preferential promotion of a Th2 cytokine-secreting profile. In exerting their antiproliferative effects, GCs influence both transcriptional and posttranscriptional events by binding their cytosolic receptor (GR), which subsequently binds the promoter region of cytokine genes on select DNA sites compatible with the GCs responsible elements (GRE) motif. In addition to direct DNA binding, GCs may also directly bind to, and hence antagonize, nuclear factors required for efficient gene expression, thereby markedly reducing transcriptional rate. The pleiotrophy of the GCs action, coupled with the diverse experimental conditions employed in assessing the GCs effects, indicate that GCs may utilize more than one mechanism in inhibiting T cell activation, and warrant careful scrutiny in assigning a mechanism by which GCs exert their antiproliferative effects. © 1998 Elsevier Science Inc.
Collapse
Affiliation(s)
- W Y Almawi
- Medical Sciences Unit, Lebanese National Council for Scientific Research, Beirut
| | | | | |
Collapse
|
120
|
Cui X, Xie Z. Protein Interaction and Na/K-ATPase-Mediated Signal Transduction. Molecules 2017; 22:molecules22060990. [PMID: 28613263 PMCID: PMC6152704 DOI: 10.3390/molecules22060990] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 02/05/2023] Open
Abstract
The Na/K-ATPase (NKA), or Na pump, is a member of the P-type ATPase superfamily. In addition to pumping ions across cell membrane, it is engaged in assembly of multiple protein complexes in the plasma membrane. This assembly allows NKA to perform many non-pumping functions including signal transduction that are important for animal physiology and disease progression. This article will focus on the role of protein interaction in NKA-mediated signal transduction, and its potential utility as target for developing new therapeutics.
Collapse
Affiliation(s)
- Xiaoyu Cui
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV 25703, USA.
| |
Collapse
|
121
|
Xu H, Zong H, Ma C, Ming X, Shang M, Li K, He X, Du H, Cao L. Epidermal growth factor receptor in glioblastoma. Oncol Lett 2017; 14:512-516. [PMID: 28693199 PMCID: PMC5494611 DOI: 10.3892/ol.2017.6221] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 03/21/2017] [Indexed: 12/11/2022] Open
Abstract
Mutations in the epidermal growth factor receptor (EGFR) are commonly occurring in glioblastoma. Enhanced activation of EGFR can occur through a variety of different mechanisms, both ligand-dependent and ligand-independent. Numerous evidence has suggested that EGFR is overexpressed in most of primary glioblastomas and some of the secondary glioblastomas and is characteristic of more aggressive glioblastoma phenotypes. Additionally, recent studies have revealed that wild-type EGFR, and to a greater extent hyper-activating EGFR mutants induced a substantial upregulation of Fyn expression. Furthermore, it was determined that Fyn expression is upregulated across a panel of patient-derived glioblastoma stem cells (GSCs) relative to normal progenitor controls. Moreover, researchers are continuously involved in elucidation of novel mechanism linking EGFR EGFR-expressing glioblastoma. The present review highlights current aspects of EGFR receptor in glioblastoma and concludes that the concept of EGFR signaling and related receptors and associated factors is evolving, however, it needs detailed evaluation for future clinical applications in cancer patients.
Collapse
Affiliation(s)
- Hongsheng Xu
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Hailiang Zong
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Chong Ma
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Xing Ming
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Ming Shang
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Kai Li
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Xiaoguang He
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Hai Du
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| | - Lei Cao
- Department of Neurosurgery, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| |
Collapse
|
122
|
Brown CW, Sridhara V, Boutz DR, Person MD, Marcotte EM, Barrick JE, Wilke CO. Large-scale analysis of post-translational modifications in E. coli under glucose-limiting conditions. BMC Genomics 2017; 18:301. [PMID: 28412930 PMCID: PMC5392934 DOI: 10.1186/s12864-017-3676-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 03/31/2017] [Indexed: 01/24/2023] Open
Abstract
Background Post-translational modification (PTM) of proteins is central to many cellular processes across all domains of life, but despite decades of study and a wealth of genomic and proteomic data the biological function of many PTMs remains unknown. This is especially true for prokaryotic PTM systems, many of which have only recently been recognized and studied in depth. It is increasingly apparent that a deep sampling of abundance across a wide range of environmental stresses, growth conditions, and PTM types, rather than simply cataloging targets for a handful of modifications, is critical to understanding the complex pathways that govern PTM deposition and downstream effects. Results We utilized a deeply-sampled dataset of MS/MS proteomic analysis covering 9 timepoints spanning the Escherichia coli growth cycle and an unbiased PTM search strategy to construct a temporal map of abundance for all PTMs within a 400 Da window of mass shifts. Using this map, we are able to identify novel targets and temporal patterns for N-terminal N α acetylation, C-terminal glutamylation, and asparagine deamidation. Furthermore, we identify a possible relationship between N-terminal N α acetylation and regulation of protein degradation in stationary phase, pointing to a previously unrecognized biological function for this poorly-understood PTM. Conclusions Unbiased detection of PTM in MS/MS proteomics data facilitates the discovery of novel modification types and previously unobserved dynamic changes in modification across growth timepoints. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3676-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Colin W Brown
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Viswanadham Sridhara
- Center for Computational Biology and Bioinformatics, The University of Texas at Austin, Austin, Texas, USA
| | - Daniel R Boutz
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA.,Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Maria D Person
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA.,College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| | - Edward M Marcotte
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA.,Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, Texas, USA.,Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Jeffrey E Barrick
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA.,Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, Texas, USA.,Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Claus O Wilke
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA. .,Center for Computational Biology and Bioinformatics, The University of Texas at Austin, Austin, Texas, USA. .,Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA.
| |
Collapse
|
123
|
Wang J, Chen R, Liu X, Shen J, Yan Y, Gao Y, Tao T, Shi J. Hck Promotes Neuronal Apoptosis Following Intracerebral Hemorrhage. Cell Mol Neurobiol 2017; 37:251-261. [PMID: 27053350 PMCID: PMC11482238 DOI: 10.1007/s10571-016-0365-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/21/2016] [Indexed: 01/04/2023]
Abstract
The hematopoietic cell kinase (Hck) is a member of the Src family protein kinases which regulates many signal transduction pathways including cell growth, proliferation, differentiation, migration, and apoptosis. However, the expression and function of Hck after intracerebral hemorrhage (ICH) are unknown. Western blot, immunohistochemistry, and immunofluorescence showed that Hck was obviously up-regulation in neurons adjacent to the hematoma after ICH. In addition, the temporary raise of Hck expression was paralleled with the expression of p53, Bax, and active caspase-3, suggesting that Hck was involved in neuronal apoptosis. Hck siRNA dramatically decrease hemin-induced expression of p53, Bax, and active caspase-3 as well as the amount of apoptotic SH-SY5Y cells in vitro. Furthermore, Hck interacted with p53. Hence, Hck might promote neuronal apoptosis via p53 signaling pathway after ICH.
Collapse
Affiliation(s)
- Jun Wang
- Department of Geriatric Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Rongrong Chen
- Department of Geriatric Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xiaojuan Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jianhong Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Yaohua Yan
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Yilu Gao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Tao Tao
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Jiansheng Shi
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
124
|
Noguchi T, Ishiba H, Honda K, Kondoh Y, Osada H, Ohno H, Fujii N, Oishi S. Synthesis of Grb2 SH2 Domain Proteins for Mirror-Image Screening Systems. Bioconjug Chem 2017; 28:609-619. [DOI: 10.1021/acs.bioconjchem.6b00692] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Taro Noguchi
- Graduate
School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Ishiba
- Graduate
School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kaori Honda
- Chemical
Biology Research Group, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Yasumitsu Kondoh
- Chemical
Biology Research Group, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Hiroyuki Osada
- Chemical
Biology Research Group, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Hiroaki Ohno
- Graduate
School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Nobutaka Fujii
- Graduate
School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shinya Oishi
- Graduate
School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
125
|
Abstract
SH2 domains first shed light on the key role of modular binding domains in cell signaling. Much of what we now know about the logic and design principles underlying cell signaling mechanisms, and how such mechanisms might have evolved, can be traced back to early work on SH2 domains. Here we briefly outline several key concepts that emerged from such studies.
Collapse
Affiliation(s)
- Bruce J Mayer
- Raymond and Beverly Sackler Laboratory of Genetics and Molecular Medicine, Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 400 Farmington Avenue, Farmington, CT, USA.
| |
Collapse
|
126
|
Abstract
The ErbB receptor family, also known as the EGF receptor family or type I receptor family, includes the epidermal growth factor (EGF) receptor (EGFR) or ErbB1/Her1, ErbB2/Her2, ErbB3/Her3, and ErbB4/Her4. Among all RTKs, EGFR was the first RTK identified and the first one linked to cancer. Thus, EGFR has also been the most intensively studied among all RTKs. ErbB receptors are activated after homodimerization or heterodimerization. The ErbB family is unique among the various groups of receptor tyrosine kinases (RTKs) in that ErbB3 has impaired kinase activity, while ErbB2 does not have a direct ligand. Therefore, heterodimerization is an important mechanism that allows the activation of all ErbB receptors in response to ligand stimulation. The activated ErbB receptors bind to many signaling proteins and stimulate the activation of many signaling pathways. The specificity and potency of intracellular signaling pathways are determined by positive and negative regulators, the specific composition of activating ligand(s), receptor dimer components, and the diverse range of proteins that associate with the tyrosine phosphorylated C-terminal domain of the ErbB receptors. ErbB receptors are overexpressed or mutated in many cancers, especially in breast cancer, ovarian cancer, and non-small cell lung cancer. The overexpression and overactivation of ErbB receptors are correlated with poor prognosis, drug resistance, cancer metastasis, and lower survival rate. ErbB receptors, especially EGFR and ErbB2 have been the primary choices as targets for developing cancer therapies.
Collapse
Affiliation(s)
- Zhixiang Wang
- Signal Transduction Research Group, Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, 835 MSB, 114 St NW, Edmonton, AB, Canada, T6G 2H7.
| |
Collapse
|
127
|
Abstract
The Src Homology 2 (SH2) domain is the prototypical protein interaction module that lies at the heart of phosphotyrosine signaling. Since its serendipitous discovery, there has been a tremendous advancement in technologies and an array of techniques available for studying SH2 domains and phosphotyrosine signaling. In this chapter, we provide a glimpse of the history of SH2 domains and describe many of the tools and techniques that have been developed along the way and discuss future directions for SH2 domain studies. We highlight the gist of each chapter in this volume in the context of: the structural biology and phosphotyrosine binding; characterizing SH2 specificity and generating prediction models; systems biology and proteomics; SH2 domains in signal transduction; and SH2 domains in disease, diagnostics, and therapeutics. Many of the individual chapters provide an in-depth approach that will allow scientists to interrogate the function and role of SH2 domains.
Collapse
Affiliation(s)
- Bernard A Liu
- Broad Institute of Harvard and MIT, 415 Main St., 5175 JJ, Cambridge, MA, 02142, USA.
| | - Kazuya Machida
- Raymond and Beverly Sackler Laboratory of Genetics and Molecular Medicine, Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 400 Farmington Ave., Farmington, CT, 06030, USA.
| |
Collapse
|
128
|
Mass-spectrometric exploration of proteome structure and function. Nature 2016; 537:347-55. [PMID: 27629641 DOI: 10.1038/nature19949] [Citation(s) in RCA: 1404] [Impact Index Per Article: 156.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 07/15/2016] [Indexed: 12/18/2022]
Abstract
Numerous biological processes are concurrently and coordinately active in every living cell. Each of them encompasses synthetic, catalytic and regulatory functions that are, almost always, carried out by proteins organized further into higher-order structures and networks. For decades, the structures and functions of selected proteins have been studied using biochemical and biophysical methods. However, the properties and behaviour of the proteome as an integrated system have largely remained elusive. Powerful mass-spectrometry-based technologies now provide unprecedented insights into the composition, structure, function and control of the proteome, shedding light on complex biological processes and phenotypes.
Collapse
|
129
|
Liang L, Zuo YF, Wu W, Zhu XQ, Yang Y. Optofluidic restricted imaging, spectroscopy and counting of nanoparticles by evanescent wave using immiscible liquids. LAB ON A CHIP 2016; 16:3007-3014. [PMID: 26984126 DOI: 10.1039/c6lc00078a] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Conventional flow cytometry (FC) suffers from the diffraction limit for the detection of nanoparticles smaller than 100 nm, whereas traditional total internal reflection (TIR) microscopy can only detect few samples near the solid-liquid interface mostly in static states. Here we demonstrate a novel on-chip optofluidic technique using evanescent wave sensing for single nanoparticle real time detection by combining hydrodynamic focusing and TIR using immiscible flows. The immiscibility of the high-index sheath flow and the low-index core flow naturally generate a smooth, flat and step-index interface that is ideal for the TIR effect, whose evanescent field can penetrate the full width of the core flow. Hydrodynamic focusing can focus on all the nanoparticles in the extreme centre of the core flow with a width smaller than 1 μm. This technique enables us to illuminate every single sample in the running core flow by the evanescent field, leaving none unaffected. Moreover, it works well for samples much smaller than the diffraction limit. We have successfully demonstrated the scattering imaging and counting of 50 nm and 100 nm Au nanoparticles and also the fluorescence imaging and counting of 200 nm beads. The effective counting speeds are estimated as 1500, 2300 and 2000 particles per second for the three types of nanoparticles, respectively. The optical scattering spectra were also measured to determine the size of individual Au nanoparticles. This provides a new technique to detect nanoparticles and we foresee its application in the detection of molecules for biomedical analyses.
Collapse
Affiliation(s)
- L Liang
- School of Physics & Technology, Wuhan University, Wuhan 430072, China.
| | | | | | | | | |
Collapse
|
130
|
Abbaspour Babaei M, Kamalidehghan B, Saleem M, Huri HZ, Ahmadipour F. Receptor tyrosine kinase (c-Kit) inhibitors: a potential therapeutic target in cancer cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:2443-59. [PMID: 27536065 PMCID: PMC4975146 DOI: 10.2147/dddt.s89114] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
c-Kit, a receptor tyrosine kinase, is involved in intracellular signaling, and the mutated form of c-Kit plays a crucial role in occurrence of some cancers. The function of c-Kit has led to the concept that inhibiting c-Kit kinase activity can be a target for cancer therapy. The promising results of inhibition of c-Kit for treatment of cancers have been observed in some cancers such as gastrointestinal stromal tumor, acute myeloid leukemia, melanoma, and other tumors, and these results have encouraged attempts toward improvement of using c-Kit as a capable target for cancer therapy. This paper presents the findings of previous studies regarding c-Kit as a receptor tyrosine kinase and an oncogene, as well as its gene targets and signaling pathways in normal and cancer cells. The c-Kit gene location, protein structure, and the role of c-Kit in normal cell have been discussed. Comprehending the molecular mechanism underlying c-Kit-mediated tumorogenesis is consequently essential and may lead to the identification of future novel drug targets. The potential mechanisms by which c-Kit induces cellular transformation have been described. This study aims to elucidate the function of c-Kit for future cancer therapy. In addition, it has c-Kit inhibitor drug properties and their functions have been listed in tables and demonstrated in schematic pictures. This review also has collected previous studies that targeted c-Kit as a novel strategy for cancer therapy. This paper further emphasizes the advantages of this approach, as well as the limitations that must be addressed in the future. Finally, although c-Kit is an attractive target for cancer therapy, based on the outcomes of treatment of patients with c-Kit inhibitors, it is unlikely that Kit inhibitors alone can lead to cure. It seems that c-Kit mutations alone are not sufficient for tumorogenesis, but do play a crucial role in cancer occurrence.
Collapse
Affiliation(s)
| | - Behnam Kamalidehghan
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology (NIGEB), Shahrak-e Pajoohesh; Medical Genetics Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Saleem
- Department of Urology; Department of Laboratory Medicine and Pathology, Masonic Cancer Center, University of Minnesota; Section of Molecular Therapeutics & Cancer Health Disparity, The Hormel Institute, Austin, MN, USA
| | - Hasniza Zaman Huri
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Clinical Investigation Centre, University Malaya Medical Centre, Lembah Pantai, Kuala Lumpur, Malaysia
| | - Fatemeh Ahmadipour
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
131
|
Non-Ligand-Induced Dimerization is Sufficient to Initiate the Signalling and Endocytosis of EGF Receptor. Int J Mol Sci 2016; 17:ijms17081200. [PMID: 27463710 PMCID: PMC5000598 DOI: 10.3390/ijms17081200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 07/13/2016] [Accepted: 07/19/2016] [Indexed: 11/17/2022] Open
Abstract
The binding of epidermal growth factor (EGF) to EGF receptor (EGFR) stimulates cell mitogenesis and survival through various signalling cascades. EGF also stimulates rapid EGFR endocytosis and its eventual degradation in lysosomes. The immediate events induced by ligand binding include receptor dimerization, activation of intrinsic tyrosine kinase and autophosphorylation. However, in spite of intensified efforts, the results regarding the roles of these events in EGFR signalling and internalization is still very controversial. In this study, we constructed a chimeric EGFR by replacing its extracellular domain with leucine zipper (LZ) and tagged a green fluorescent protein (GFP) at its C-terminus. We showed that the chimeric LZ-EGFR-GFP was constitutively dimerized. The LZ-EGFR-GFP dimer autophosphorylated each of its five well-defined C-terminal tyrosine residues as the ligand-induced EGFR dimer does. Phosphorylated LZ-EGFR-GFP was localized to both the plasma membrane and endosomes, suggesting it is capable of endocytosis. We also showed that LZ-EGFR-GFP activated major signalling proteins including Src homology collagen-like (Shc), extracellular signal-regulated kinase (ERK) and Akt. Moreover, LZ-EGFR-GFP was able to stimulate cell proliferation. These results indicate that non-ligand induced dimerization is sufficient to activate EGFR and initiate cell signalling and EGFR endocytosis. We conclude that receptor dimerization is a critical event in EGF-induced cell signalling and EGFR endocytosis.
Collapse
|
132
|
Evolution of domain-peptide interactions to coadapt specificity and affinity to functional diversity. Proc Natl Acad Sci U S A 2016; 113:E3862-71. [PMID: 27317745 DOI: 10.1073/pnas.1518469113] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Evolution of complexity in eukaryotic proteomes has arisen, in part, through emergence of modular independently folded domains mediating protein interactions via binding to short linear peptides in proteins. Over 30 years, structural properties and sequence preferences of these peptides have been extensively characterized. Less successful, however, were efforts to establish relationships between physicochemical properties and functions of domain-peptide interactions. To our knowledge, we have devised the first strategy to exhaustively explore the binding specificity of protein domain-peptide interactions. We applied the strategy to SH3 domains to determine the properties of their binding peptides starting from various experimental data. The strategy identified the majority (∼70%) of experimentally determined SH3 binding sites. We discovered mutual relationships among binding specificity, binding affinity, and structural properties and evolution of linear peptides. Remarkably, we found that these properties are also related to functional diversity, defined by depth of proteins within hierarchies of gene ontologies. Our results revealed that linear peptides evolved to coadapt specificity and affinity to functional diversity of domain-peptide interactions. Thus, domain-peptide interactions follow human-constructed gene ontologies, which suggest that our understanding of biological process hierarchies reflect the way chemical and thermodynamic properties of linear peptides and their interaction networks, in general, have evolved.
Collapse
|
133
|
Xie L, Xie Z, Huang L, Fan Q, Luo S, Huang J, Deng X, Xie Z, Zeng T, Zhang Y, Wang S. Avian reovirus σA and σNS proteins activate the phosphatidylinositol 3-kinase-dependent Akt signalling pathway. Arch Virol 2016; 161:2243-8. [PMID: 27233800 DOI: 10.1007/s00705-016-2908-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/19/2016] [Indexed: 11/26/2022]
Abstract
The present study was conducted to identify avian reovirus (ARV) proteins that can activate the phosphatidylinositol 3-kinase (PI3K)-dependent Akt pathway. Based on ARV protein amino acid sequence analysis, σA, σNS, μA, μB and μNS were identified as putative proteins capable of mediating PI3K/Akt pathway activation. The recombinant plasmids σA-pcAGEN, σNS-pcAGEN, μA-pcAGEN, μB-pcAGEN and μNS-pcAGEN were constructed and used to transfect Vero cells, and the expression levels of the corresponding genes were quantified by immunofluorescence and Western blot analysis. Phosphorylated Akt (P-Akt) levels in the transfected cells were measured by flow cytometry and Western blot analysis. The results showed that the σA, σNS, μA, μB and μNS genes were expressed in Vero cells. σA-expressing and σNS-expressing cells had higher P-Akt levels than negative control cells, pcAGEN-expressing cells and cells designed to express other proteins (i.e., μA, μB and μNS). Pre-treatment with the PI3K inhibitor LY294002 inhibited Akt phosphorylation in σA- and σNS-expressing cells. These results indicate that the σA and σNS proteins can activate the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Liji Xie
- Department of Biotechnology, Guangxi Key Laboratory of Animal Epidemic Etiology and Diagnostics, Guangxi Veterinary Research Institute, 51 Youai North Road, Nanning, 530001, China
| | - Zhixun Xie
- Department of Biotechnology, Guangxi Key Laboratory of Animal Epidemic Etiology and Diagnostics, Guangxi Veterinary Research Institute, 51 Youai North Road, Nanning, 530001, China.
| | - Li Huang
- Department of Biotechnology, Guangxi Key Laboratory of Animal Epidemic Etiology and Diagnostics, Guangxi Veterinary Research Institute, 51 Youai North Road, Nanning, 530001, China
| | - Qing Fan
- Department of Biotechnology, Guangxi Key Laboratory of Animal Epidemic Etiology and Diagnostics, Guangxi Veterinary Research Institute, 51 Youai North Road, Nanning, 530001, China
| | - Sisi Luo
- Department of Biotechnology, Guangxi Key Laboratory of Animal Epidemic Etiology and Diagnostics, Guangxi Veterinary Research Institute, 51 Youai North Road, Nanning, 530001, China
| | - Jiaoling Huang
- Department of Biotechnology, Guangxi Key Laboratory of Animal Epidemic Etiology and Diagnostics, Guangxi Veterinary Research Institute, 51 Youai North Road, Nanning, 530001, China
| | - Xianwen Deng
- Department of Biotechnology, Guangxi Key Laboratory of Animal Epidemic Etiology and Diagnostics, Guangxi Veterinary Research Institute, 51 Youai North Road, Nanning, 530001, China
| | - Zhiqin Xie
- Department of Biotechnology, Guangxi Key Laboratory of Animal Epidemic Etiology and Diagnostics, Guangxi Veterinary Research Institute, 51 Youai North Road, Nanning, 530001, China
| | - Tingting Zeng
- Department of Biotechnology, Guangxi Key Laboratory of Animal Epidemic Etiology and Diagnostics, Guangxi Veterinary Research Institute, 51 Youai North Road, Nanning, 530001, China
| | - Yanfang Zhang
- Department of Biotechnology, Guangxi Key Laboratory of Animal Epidemic Etiology and Diagnostics, Guangxi Veterinary Research Institute, 51 Youai North Road, Nanning, 530001, China
| | - Sheng Wang
- Department of Biotechnology, Guangxi Key Laboratory of Animal Epidemic Etiology and Diagnostics, Guangxi Veterinary Research Institute, 51 Youai North Road, Nanning, 530001, China
| |
Collapse
|
134
|
Yuan YL, Lin BQ, Zhang CF, Cui LL, Ruan SX, Yang ZL, Li F, Ji D. Timosaponin B-II Ameliorates Palmitate-Induced Insulin Resistance and Inflammation via IRS-1/PI3K/Akt and IKK/NF-[Formula: see text]B Pathways. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 44:755-69. [PMID: 27222060 DOI: 10.1142/s0192415x16500415] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This study aimed to investigate the effect of timosaponin B-II (TB-II) on palmitate (PA)-induced insulin resistance and inflammation in HepG2 cells, and probe the potential mechanisms. TB-II, a main ingredient of the traditional Chinese medicine Anemarrhena asphodeloides Bunge, notably ameliorated PA-induced insulin resistance and inflammation, and significantly improved cell viability, decreased PA-induced production of tumor necrosis factor-[Formula: see text] (TNF-[Formula: see text]) and interleukin-6 (IL-6) levels. Further, TB-II treatment notably decreased malondialdehyde (MDA) and lactate dehydrogenase (LDH) levels, and improved superoxide dismutase (SOD) and nitric oxide (NO). TB-II also reduced HepG2 cells apoptosis. Insulin receptor substrate-1 (IRS1)/phosphatidylinositol 3-kinase (PI3K)/Akt and inhibitor of nuclear factor [Formula: see text]-B kinase (IKK)/NF-[Formula: see text]B pathways-related proteins, and IKK[Formula: see text], p65 phosphorylation, serine phosphorylation of insulin receptor substrate-1 (IRS-1) at S307, tyrosine phosphorylation of IRS-1, and Akt activation were determined by Western blot. Compared to model group, TB-II significantly downregulated the expression of p-NF-[Formula: see text]Bp65, p-IKK[Formula: see text], p-IRS-1, p-PI3K and p-Akt. TB-II is a promising potential agent for the management of palmitate-induced insulin resistance and inflammation, which might be via IR/IRS-1/PI3K/Akt and IKK/NF-[Formula: see text]B pathways.
Collapse
Affiliation(s)
- Yong-Liang Yuan
- * State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Bao-Qin Lin
- † Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Chun-Feng Zhang
- * State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Ling-Ling Cui
- * State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Shi-Xia Ruan
- * State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Zhong-Lin Yang
- * State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Fei Li
- * State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - De Ji
- ‡ College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| |
Collapse
|
135
|
Frequent mutations in acetylation and ubiquitination sites suggest novel driver mechanisms of cancer. Genome Med 2016; 8:55. [PMID: 27175787 PMCID: PMC4864925 DOI: 10.1186/s13073-016-0311-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 04/19/2016] [Indexed: 12/14/2022] Open
Abstract
Background Discovery of cancer drivers is a major goal of cancer research. Driver genes and pathways are often predicted using mutation frequency, assuming that statistically significant recurrence of specific somatic mutations across independent samples indicates their importance in cancer. However, many mutations, including known cancer drivers, are not observed at high frequency. Fortunately, abundant information is available about functional “active sites” in proteins that can be integrated with mutations to predict cancer driver genes, even based on low frequency mutations. Further, considering active site information predicts detailed biochemical mechanisms impacted by the mutations. Post-translational modifications (PTMs) are active sites that are regulatory switches in proteins and pathways. We analyzed acetylation and ubiquitination, two important PTM types often involved in chromatin organization and protein degradation, to find proteins that are significantly affected by tumor somatic mutations. Methods We performed computational analyses of acetylation and ubiquitination sites in a pan-cancer dataset of 3200 tumor samples from The Cancer Genome Atlas (TCGA). These analyses were targeted at different levels of biological organization including individual genes, pathway annotated gene sets, and protein-protein interaction networks. Results Acetylation and ubiquitination site mutations are enriched in cancer with significantly stronger evolutionary conservation and accumulation in protein domains. Gene-focused analysis with the ActiveDriver method reveals significant co-occurrences of acetylation and ubiquitination PTMs and mutation hotspots in known oncoproteins (TP53, AKT1, IDH1) and highlights candidate cancer driver genes with PTM-related mechanisms (e.g. several histone proteins and the splicing factor SF3B1). Pathway analysis shows that PTM mutations in acetylation and ubiquitination sites accumulate in cancer-related processes such as cell cycle, apoptosis, chromatin regulation, and metabolism. Integrated mutation analysis of clinical information and protein interaction networks suggests that many PTM-specific mutations associate with decreased patient survival. Conclusions Mutation analysis of acetylation and ubiquitination PTM sites reveals their importance in cancer. As PTM networks are increasingly mapped and related enzymes are often druggable, deeper investigation of specific associated mutations may lead to the discovery of treatment-relevant cellular mechanisms. Electronic supplementary material The online version of this article (doi:10.1186/s13073-016-0311-2) contains supplementary material, which is available to authorized users.
Collapse
|
136
|
Rorsman C, Tsioumpekou M, Heldin CH, Lennartsson J. The Ubiquitin Ligases c-Cbl and Cbl-b Negatively Regulate Platelet-derived Growth Factor (PDGF) BB-induced Chemotaxis by Affecting PDGF Receptor β (PDGFRβ) Internalization and Signaling. J Biol Chem 2016; 291:11608-18. [PMID: 27048651 DOI: 10.1074/jbc.m115.705814] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Indexed: 11/06/2022] Open
Abstract
Protein ubiquitination controls protein stability and subcellular localization of tyrosine kinase receptors, hence affecting signaling both quantitatively and qualitatively. In this report, we demonstrate that, after ligand stimulation, the PDGF β receptor (PDGFRβ) becomes ubiquitinated in a manner requiring both the c-Cbl and Cbl-b ubiquitin ligases. Simultaneous depletion of c-Cbl and Cbl-b resulted in reduced ligand-induced PDGFRβ clearance from the cell surface because of reduced endocytosis of the receptor. Cbl-b formed a complex with c-Cbl, as well as with the PDGFRβ, in response to PDGF-BB stimulation. We were unable to find a direct interaction between the receptor and c-Cbl, raising the possibility that Cbl-b is necessary for c-Cbl to interact with PDGFRβ. Phosphorylated Tyr-1021 in PDGFRβ was the primary interaction site for Cbl-b, with some contribution from Tyr-1009. Depletion of c-Cbl and Cbl-b led to an increased ligand-induced tyrosine phosphorylation of the receptor. Several tyrosine residues with elevated phosphorylation (i.e. Tyr-579, Tyr-581, Tyr-1009, and Tyr-1021) have previously been shown to interact with Src kinases and PLCγ. Indeed, in cells depleted of c-Cbl and Cbl-b, both Src and PLCγ phosphorylation were enhanced, whereas activation of other pathways, such as Erk1/2 MAP kinase and Akt, were not affected. In addition, Stat3 phosphorylation, which has been connected to Src activity, was also elevated in cells lacking c-Cbl and Cbl-b. Functionally, we found that cells depleted of c-Cbl and Cbl-b were more prone to migrate toward PDGF-BB, whereas no reproducible effect on cell proliferation could be observed. In conclusion, internalization as well as signaling via PDGFRβ are controlled by ubiquitination.
Collapse
Affiliation(s)
- Charlotte Rorsman
- From the Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, 75124 Uppsala, Sweden
| | - Maria Tsioumpekou
- From the Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, 75124 Uppsala, Sweden
| | - Carl-Henrik Heldin
- From the Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, 75124 Uppsala, Sweden
| | - Johan Lennartsson
- From the Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, 75124 Uppsala, Sweden
| |
Collapse
|
137
|
Hansen S, Tremmel D, Madhurantakam C, Reichen C, Mittl PRE, Plückthun A. Structure and Energetic Contributions of a Designed Modular Peptide-Binding Protein with Picomolar Affinity. J Am Chem Soc 2016; 138:3526-32. [DOI: 10.1021/jacs.6b00099] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Simon Hansen
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Dirk Tremmel
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Chaithanya Madhurantakam
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Christian Reichen
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Peer R. E. Mittl
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| |
Collapse
|
138
|
Chen H, Wang D, Liu Y. SASH1 inhibits cervical cancer cell proliferation and invasion by suppressing the FAK pathway. Mol Med Rep 2016; 13:3613-8. [PMID: 26935246 DOI: 10.3892/mmr.2016.4946] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 11/06/2015] [Indexed: 11/05/2022] Open
Abstract
SAM and SH3 domain containing 1 (SASH1), a member of the SLY-family of signal adapter proteins, is a candidate tumor suppressor in several types of cancer. However, the role of SASH1 in cervical cancer remains to be elucidated. Therefore, in the present study, the role of SASH1 in cervical cancer and the underlying mechanism was investigated. Cell proliferation was detected by the MTT assay. Cell invasion was measured by Transwell assay. The mRNA expression levels of SASH1, matrix metalloproteinase (MMP)‑2 and MMP‑9 were determined by reverse transcription quantitative polymerase chain reaction. The protein expression levels of SASH1, MMP‑2, MMP‑9 and focal adhesion kinase (FAK) were determined by western blot analysis. The results demonstrated that SASH1 was downregulated in cervical cancer tissues and cell lines. Subsequently, a vector that overexpresses SASH1 was constructed. Overexpression of SASH1 was found to significantly inhibit cervical cancer cell proliferation and invasion, and also significantly reduce the expression of MMP‑2 and MMP‑9 in cancer cells. In addition, SASH1 modulated the FAK signaling pathway. Overexpression of SASH1 suppressed the expression of FAK in cancer cells. Taken together, the results suggested that SASH1 inhibits cervical cancer cell proliferation and invasion by suppressing the FAK pathway.
Collapse
Affiliation(s)
- Hui Chen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Dongliang Wang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Yuling Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| |
Collapse
|
139
|
Affiliation(s)
- Steven A Stacker
- a Tumour Angiogenesis Program, Peter MacCallum Cancer Centre , East Melbourne , VIC , Australia
- b Sir Peter MacCallum Department of Oncology , University of Melbourne , VIC , Australia , and
- c Department of Surgery , Royal Melbourne Hospital, University of Melbourne , Parkville , VIC , Australia
| |
Collapse
|
140
|
Leng G, Song K. Direct interaction of Ste11 and Mkk1/2 through Nst1 integrates high-osmolarity glycerol and pheromone pathways to the cell wall integrity MAPK pathway. FEBS Lett 2016; 590:148-60. [PMID: 26787465 DOI: 10.1002/1873-3468.12039] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 11/28/2015] [Accepted: 12/03/2015] [Indexed: 11/11/2022]
Abstract
Coordination and cross talks of MAPK pathways are critical for signaling efficiency, but their mechanisms are not well understood. Slt2, the MAP kinase of cell wall integrity pathway (CWI), is activated by heat stress even in the absence of upstream components of this pathway, suggesting a supplementary input for Slt2 activation. Here, we identify a new interaction of Ste11 and Mkk1, mediated by Nst1 that connects the high-osmolarity glycerol and pheromone pathways directly to CWI pathway in response to heat and pheromone. We suggest that Ser(407) and Thr(411) are novel residues of Mkk1 activated by these MAPK pathways.
Collapse
Affiliation(s)
- Gang Leng
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Kiwon Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| |
Collapse
|
141
|
Full coding hepatitis E virus genotype 3 genome amplification method. J Virol Methods 2016; 230:18-23. [PMID: 26784284 PMCID: PMC7172825 DOI: 10.1016/j.jviromet.2016.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 01/12/2016] [Accepted: 01/14/2016] [Indexed: 02/07/2023]
Abstract
A new amplification method for the complete HEV coding genome was described. HEV genome organization and function were reviewed. Polymorphic amino acids of each HEV genome region were analyzed in reference sequences. Complete coding HEV sequence of an immunocompromised patient with acute infection was analyzed.
Hepatitis E virus (HEV) genotype 3 produces zoonotic infection associated with the consumption of infected animals. HEV infections can become chronic in immunocompromised (IC) patients. The viral genome has three well defined open reading frames (ORF1, ORF2 and ORF3) within which various domains and functions have been described. This paper (i) describes a new method of complete sequencing of the HEV coding region through overlapping PCR systems, (ii) establishes a consensus sequence and polymorphic positions (PP) for each domain, and (iii) analyzes the complete coding sequence of an IC patient. With regard to the consensus, a high percentage of PP was observed in protease (PP = 19%) and the X domain (PP = 22%) within ORF1, the N-terminal region of the S domain (PP = 22%) in ORF2, and the P1 (PP = 35%) and P2 (PP = 25%) domains in ORF3. In contrast, the ORF1 Y, ORF2 S, ORF2 M and ORF3 D1 domains were conserved in the reference sequences (0.40, 1, 0.70 and 0% of PP, respectively). The sequence from the IC patient had more mutations in the RpRp (D1235G, Q1242R, S1454T, V1480I, I1502 V, K1511R, G1373 V, E1442D, V1693 M), the terminal ORF2 S- domain (F10L, S26T, G36S, S70P, A105 V, I113 V), the X domain (T938 M, T856 V, S898A) and the helicase (S1014N, S975T, Q1133 K).
Collapse
|
142
|
Wang Z. Transactivation of Epidermal Growth Factor Receptor by G Protein-Coupled Receptors: Recent Progress, Challenges and Future Research. Int J Mol Sci 2016; 17:ijms17010095. [PMID: 26771606 PMCID: PMC4730337 DOI: 10.3390/ijms17010095] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/06/2016] [Accepted: 01/07/2016] [Indexed: 02/06/2023] Open
Abstract
Both G protein-coupled receptors (GPCRs) and receptor-tyrosine kinases (RTKs) regulate large signaling networks, control multiple cell functions and are implicated in many diseases including various cancers. Both of them are also the top therapeutic targets for disease treatment. The discovery of the cross-talk between GPCRs and RTKs connects these two vast signaling networks and complicates the already complicated signaling networks that regulate cell signaling and function. In this review, we focus on the transactivation of epidermal growth factor receptor (EGFR), a subfamily of RTKs, by GPCRs. Since the first report of EGFR transactivation by GPCR, significant progress has been made including the elucidation of the mechanisms underlying the transactivation. Here, we first provide a basic picture for GPCR, EGFR and EGFR transactivation by GPCR. We then discuss the progress made in the last five years and finally provided our view of the future challenge and future researches needed to overcome these challenges.
Collapse
Affiliation(s)
- Zhixiang Wang
- The Department of Medical Genetics and Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
143
|
Sample Preparation for Mass Spectrometry Analysis of Protein-Protein Interactions in Cancer Cell Lines and Tissues. Methods Mol Biol 2016; 1458:339-47. [PMID: 27581032 DOI: 10.1007/978-1-4939-3801-8_23] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A precisely controlled network of protein-protein interactions constitutes the basis for functional signaling pathways. This equilibrium is more often than not disrupted in cancer cells, by the aberrant expression or activation of oncogenic proteins. Therefore, the analysis of protein interaction networks in cancer cells has become crucial to expand our comprehension of the molecular underpinnings of tumor formation and progression. This protocol describes a sample preparation method for the analysis of signaling complexes by mass spectrometry (MS), following the affinity purification of a protein of interest from a cancer cell line or a solid tumor. In particular, we provide a spin tip-based protease digestion procedure that offers a more rapid and controlled alternative to other gel-based and gel-free methods. This sample preparation protocol represents a useful strategy to identify protein interactions and to gain insight into the molecular mechanisms that contribute to a given cancer phenotype.
Collapse
|
144
|
Jacobsen FA, Hulst C, Bäckström T, Koleske AJ, Andersson Å. Arg Deficiency Does not Influence the Course of Myelin Oligodendrocyte Glycoprotein (MOG35-55)-induced Experimental Autoimmune Encephalomyelitis. ACTA ACUST UNITED AC 2016; 7. [PMID: 34527426 PMCID: PMC8439389 DOI: 10.4172/2155-9899.1000420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Background Inhibition of Abl kinases has an ameliorating effect on the rodent model for multiple sclerosis, experimental autoimmune encephalomyelitis, and arrests lymphocyte activation. The family of Abl kinases consists of the Abl1/Abl and Abl2/Arg tyrosine kinases. While the Abl kinase has been extensively studied in immune activation, roles for Arg are incompletely characterized. To investigate the role for Arg in experimental autoimmune encephalomyelitis, we studied disease development in Arg-/- mice. Methods Arg-/- and Arg+/+ mice were generated from breeding of Arg+/- mice on the C57BL/6 background. Mice were immunized with the myelin oligodendrocyte glycoprotein (MOG)35-55 peptide and disease development recorded. Lymphocyte phenotypes of wild type Arg+/+ and Arg-/- mice were studied by in vitro stimulation assays and flow cytometry. Results The breeding of Arg+/+ and Arg-/- mice showed skewing in the frequency of born Arg-/- mice. Loss of Arg function did not affect development of experimental autoimmune encephalomyelitis, but reduced the number of splenic B-cells in Arg-/- mice following immunization with MOG peptide. Conclusions Development of MOG-induced experimental autoimmune encephalomyelitis is not dependent on Arg, but Arg plays a role for the number of B cells in immunized mice. This might suggest a novel role for the Arg kinase in B-cell trafficking or regulation. Furthermore, the results suggest that Arg is important for normal embryonic development.
Collapse
Affiliation(s)
- Freja Aksel Jacobsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk A/S, Gentofte, Denmark
| | - Camilla Hulst
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk A/S, Gentofte, Denmark
| | | | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven CT 06520, USA
| | - Åsa Andersson
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
145
|
Nieto L, Tharun IM, Balk M, Wienk H, Boelens R, Ottmann C, Milroy LG, Brunsveld L. Estrogen Receptor Folding Modulates cSrc Kinase SH2 Interaction via a Helical Binding Mode. ACS Chem Biol 2015; 10:2624-32. [PMID: 26352092 DOI: 10.1021/acschembio.5b00568] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The estrogen receptors (ERs) feature, next to their transcriptional role, important nongenomic signaling actions, with emerging clinical relevance. The Src Homology 2 (SH2) domain mediated interaction between cSrc kinase and ER plays a key role in this; however the molecular determinants of this interaction have not been elucidated. Here, we used phosphorylated ER peptide and semisynthetic protein constructs in a combined biochemical and structural study to, for the first time, provide a quantitative and structural characterization of the cSrc SH2-ER interaction. Fluorescence polarization experiments delineated the SH2 binding motif in the ER sequence. Chemical shift perturbation analysis by nuclear magnetic resonance (NMR) together with molecular dynamics (MD) simulations allowed us to put forward a 3D model of the ER-SH2 interaction. The structural basis of this protein-protein interaction has been compared with that of the high affinity SH2 binding sequence GpYEEI. The ER features a different binding mode from that of the "two-pronged plug two-hole socket" model in the so-called specificity determining region. This alternative binding mode is modulated via the folding of ER helix 12, a structural element directly C-terminal of the key phosphorylated tyrosine. The present findings provide novel molecular entries for understanding nongenomic ER signaling and targeting the corresponding disease states.
Collapse
Affiliation(s)
- Lidia Nieto
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
of Complex Molecular Systems, Eindhoven University of Technology, 5612AZ Eindhoven, The Netherlands
| | - Inga M. Tharun
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
of Complex Molecular Systems, Eindhoven University of Technology, 5612AZ Eindhoven, The Netherlands
| | - Mark Balk
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
of Complex Molecular Systems, Eindhoven University of Technology, 5612AZ Eindhoven, The Netherlands
| | - Hans Wienk
- Bijvoet
Center for Biomolecular Research, NMR Spectroscopy Utrecht University, 3584CH Utrecht, The Netherlands
| | - Rolf Boelens
- Bijvoet
Center for Biomolecular Research, NMR Spectroscopy Utrecht University, 3584CH Utrecht, The Netherlands
| | - Christian Ottmann
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
of Complex Molecular Systems, Eindhoven University of Technology, 5612AZ Eindhoven, The Netherlands
| | - Lech-Gustav Milroy
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
of Complex Molecular Systems, Eindhoven University of Technology, 5612AZ Eindhoven, The Netherlands
| | - Luc Brunsveld
- Laboratory
of Chemical Biology, Department of Biomedical Engineering and Institute
of Complex Molecular Systems, Eindhoven University of Technology, 5612AZ Eindhoven, The Netherlands
| |
Collapse
|
146
|
Cao Z, Lu C. Quantitative Detection of Nucleocytoplasmic Transport of Native Proteins in Single Cells. Methods Mol Biol 2015; 1346:239-52. [PMID: 26542726 DOI: 10.1007/978-1-4939-2987-0_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
The detection of protein translocation (i.e., the movement of intracellular proteins among various subcellular compartments) conventionally relies on imaging and subcellular-fractionation-based techniques that do not generate information on a large cell population with single-cell resolution. Although special flow cytometric tools such as imaging flow cytometry may generate single-cell data on processes such as nucleocytoplasmic transport, such equipment is expensive (thus has limited accessibility) and has low throughput for examining cells due to the reliance on high-speed imaging. Here we describe a protocol for detecting translocation of native proteins using a common flow cytometer which detects fluorescence intensity without imaging. We conduct chemical release of cytosolic proteins and fluorescence immunostaining of a targeted protein. The detected fluorescence intensity is quantitatively correlated to the cytosolic/nuclear localization of the protein at the single cell level. Our technique provides a simple route for studying nucleocytoplasmic transport with single-cell resolution using common flow cytometers.
Collapse
Affiliation(s)
- Zhenning Cao
- School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Chang Lu
- Department of Chemical Engineering, Virginia Tech, Suite 235 Goodwin Hall, 635 Prices Fork Road, Blacksburg, VA, 24061, USA.
| |
Collapse
|
147
|
Taherian A, Haas TA, Davoodabadi A. Substrate-Dependent Activity of ERK and MEK Proteins in Breast Cancer (MCF7), and Kidney Embryonic (Hek-293) Cell Lines, Cultured on Different Substrates. IRANIAN JOURNAL OF CANCER PREVENTION 2015; 8:e3909. [PMID: 26634110 PMCID: PMC4667236 DOI: 10.17795/ijcp-3909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 10/04/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Breast cancer has been one of the most common types of cancer, as the leading cause of women death in world. Breast cancer has known as a heterogenic disease that the clinical path in different patients would be very different. Since the current classification has not covered the diverse clinical course of breast cancer, lots of efforts has done to find new biological markers. Integrins are hetero dimmer proteins of α and β subunits on cell membrane. After binding to extra cellular matrix (ECM), integrins activate MAPK pathway that regulated different activities like survival, differentiation, migration, immunologic response. The interaction of integrins and ECM have a key role in cancer cell activities like survival and metastasis. OBJECTIVES In this study the expression of αvβ3 integrin, substrate -dependent morphology and ERK and p-ERK activation was compared in MCF7 and Hek-293 cells lines. MATERIALS AND METHODS The expression of αvβ3 integrin was assayed by flow cytometry. These cell lines were cultured on pre-covered plates with fibronectin (FN), fibrinogen (Fg) or collagen (Col) and the expression of ERK and p-ERK proteins was assessed in attached and free cells for each substrate after 1 hour incubation. The morphology of the cells have examined under an inverted phase contrast microscope at 15 min, 1 hour, 3 hours, 5 hours and 1 day of incubatioon. RESULTS Different substrate induced the expression ERK or p-ERK differently in the two cell lines. In MCF7 cells, substrates induced the expression of ERK in all the attached cells but free cells in BSA, collagen and Fg showed a lower expression of ERK. In comparison with Hek-293 cells althought all the attached cells have expressed ERK peotein but only free cells in collagen plates showed the expression of ERK. None of the cell lines has shown any expression of ERK and p-ERK in attached or free cells except for the Hek-293 free cells in collagen platees that have shown a weak signal for p-ERK. CONCLUSIONS Overall the breast cancer cell lines MCF7 and Hek-293 cells have differently responded on similar substrates regarding morpology or ERK and MEK expressions.
Collapse
Affiliation(s)
- Aliakbar Taherian
- Kashan Gametogenesis Research Centre, Kashan University of Medical Sciences, Kashan, IR Iran
| | - Thomas A Haas
- Department of Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | | |
Collapse
|
148
|
Shi C, Deng C. Immobilized metal ion affinity chromatography ZipTip pipette tip with polydopamine modification and Ti 4+ immobilization for selective enrichment and isolation of phosphopeptides. Talanta 2015; 143:464-468. [DOI: 10.1016/j.talanta.2015.05.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/14/2015] [Accepted: 05/18/2015] [Indexed: 12/22/2022]
|
149
|
Insights into Substrate Specificity of NlpC/P60 Cell Wall Hydrolases Containing Bacterial SH3 Domains. mBio 2015; 6:e02327-14. [PMID: 26374125 PMCID: PMC4600125 DOI: 10.1128/mbio.02327-14] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bacterial SH3 (SH3b) domains are commonly fused with papain-like Nlp/P60 cell wall hydrolase domains. To understand how the modular architecture of SH3b and NlpC/P60 affects the activity of the catalytic domain, three putative NlpC/P60 cell wall hydrolases were biochemically and structurally characterized. These enzymes all have γ-d-Glu-A2pm (A2pm is diaminopimelic acid) cysteine amidase (or dl-endopeptidase) activities but with different substrate specificities. One enzyme is a cell wall lysin that cleaves peptidoglycan (PG), while the other two are cell wall recycling enzymes that only cleave stem peptides with an N-terminal l-Ala. Their crystal structures revealed a highly conserved structure consisting of two SH3b domains and a C-terminal NlpC/P60 catalytic domain, despite very low sequence identity. Interestingly, loops from the first SH3b domain dock into the ends of the active site groove of the catalytic domain, remodel the substrate binding site, and modulate substrate specificity. Two amino acid differences at the domain interface alter the substrate binding specificity in favor of stem peptides in recycling enzymes, whereas the SH3b domain may extend the peptidoglycan binding surface in the cell wall lysins. Remarkably, the cell wall lysin can be converted into a recycling enzyme with a single mutation. Peptidoglycan is a meshlike polymer that envelops the bacterial plasma membrane and bestows structural integrity. Cell wall lysins and recycling enzymes are part of a set of lytic enzymes that target covalent bonds connecting the amino acid and amino sugar building blocks of the PG network. These hydrolases are involved in processes such as cell growth and division, autolysis, invasion, and PG turnover and recycling. To avoid cleavage of unintended substrates, these enzymes have very selective substrate specificities. Our biochemical and structural analysis of three modular NlpC/P60 hydrolases, one lysin, and two recycling enzymes, show that they may have evolved from a common molecular architecture, where the substrate preference is modulated by local changes. These results also suggest that new pathways for recycling PG turnover products, such as tracheal cytotoxin, may have evolved in bacteria in the human gut microbiome that involve NlpC/P60 cell wall hydrolases.
Collapse
|
150
|
Computational investigation of molecular mechanism and neuropathological implications in Huntington disease. Mol Cell Biochem 2015; 409:1-11. [DOI: 10.1007/s11010-015-2462-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 05/23/2015] [Indexed: 12/31/2022]
|