201
|
Hyper-phosphorylation of nsp2-related proteins of porcine reproductive and respiratory syndrome virus. Virology 2020; 543:63-75. [PMID: 32174300 PMCID: PMC7112050 DOI: 10.1016/j.virol.2020.01.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/31/2020] [Accepted: 01/31/2020] [Indexed: 01/07/2023]
Abstract
Viruses exploit phosphorylation of both viral and host proteins to support viral replication. In this study, we demonstrate that porcine reproductive and respiratory syndrome virus replicase nsp2, and two nsp2-related −2/−1 frameshifting products, nsp2TF and nsp2N, are hyper-phosphorylated. By mapping phosphorylation sites, we subdivide an extended, previously uncharacterized region, located between the papain-like protease-2 (PLP2) domain and frameshifting site, into three distinct domains. These domains include two large hypervariable regions (HVR) with putative intrinsically disordered structures, separated by a conserved and partly structured interval domain that we defined as the inter-HVR conserved domain (IHCD). Abolishing phosphorylation of the inter-species conserved residue serine918, which is located within the IHCD region, abrogates accumulation of viral genomic and subgenomic RNAs and recombinant virus production. Our study reveals the biological significance of phosphorylation events in nsp2-related proteins, emphasizes pleiotropic functions of nsp2-related proteins in the viral life cycle, and presents potential links to pathogenesis.
Collapse
|
202
|
Solleder M, Guillaume P, Racle J, Michaux J, Pak HS, Müller M, Coukos G, Bassani-Sternberg M, Gfeller D. Mass Spectrometry Based Immunopeptidomics Leads to Robust Predictions of Phosphorylated HLA Class I Ligands. Mol Cell Proteomics 2020; 19:390-404. [PMID: 31848261 PMCID: PMC7000122 DOI: 10.1074/mcp.tir119.001641] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/06/2019] [Indexed: 12/19/2022] Open
Abstract
The presentation of peptides on class I human leukocyte antigen (HLA-I) molecules plays a central role in immune recognition of infected or malignant cells. In cancer, non-self HLA-I ligands can arise from many different alterations, including non-synonymous mutations, gene fusion, cancer-specific alternative mRNA splicing or aberrant post-translational modifications. Identifying HLA-I ligands remains a challenging task that requires either heavy experimental work for in vivo identification or optimized bioinformatics tools for accurate predictions. To date, no HLA-I ligand predictor includes post-translational modifications. To fill this gap, we curated phosphorylated HLA-I ligands from several immunopeptidomics studies (including six newly measured samples) covering 72 HLA-I alleles and retrieved a total of 2,066 unique phosphorylated peptides. We then expanded our motif deconvolution tool to identify precise binding motifs of phosphorylated HLA-I ligands. Our results reveal a clear enrichment of phosphorylated peptides among HLA-C ligands and demonstrate a prevalent role of both HLA-I motifs and kinase motifs on the presentation of phosphorylated peptides. These data further enabled us to develop and validate the first predictor of interactions between HLA-I molecules and phosphorylated peptides.
Collapse
Affiliation(s)
- Marthe Solleder
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Philippe Guillaume
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Julien Racle
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Justine Michaux
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Department of Oncology, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland
| | - Hui-Song Pak
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Department of Oncology, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland
| | - Markus Müller
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Department of Oncology, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Department of Oncology, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland.
| | - David Gfeller
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| |
Collapse
|
203
|
Recent developments of chalcones as potential antibacterial agents in medicinal chemistry. Eur J Med Chem 2020; 187:111980. [DOI: 10.1016/j.ejmech.2019.111980] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 12/31/2022]
|
204
|
Ferré-Dolcet L, Rodríguez-Gil JE, Yeste M, Rigau T, Rivera Del Alamo MM. Tyrosine phosphorylation is not a relevant mechanism to modulate aquaporin 2 activity in gestational queen endometrium and placenta. Reprod Domest Anim 2020; 55:448-453. [PMID: 31951059 DOI: 10.1111/rda.13636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/09/2020] [Indexed: 11/30/2022]
Abstract
Aquaporins have been shown to be regulated by phosphorylation of serine residues, but the possible role of tyrosine residues phosphorylation has not been evaluated. Changes in the localization of aquaporin 2 (AQP2) in the queen endometrium have been related to serum progesterone levels. The aim of this study was to determine whether these AQP2-localization changes are mediated by variations in its tyrosine phosphorylation levels. Twelve queens were included in the study and divided into (a) non-macroscopically pregnant with low levels of progesterone; (b) non-macroscopically pregnant with high levels of progesterone; (c) 30 days of pregnancy; and (d) 60 days of pregnancy. Samples from endometrium and placental transference zone were obtained, immunoprecipitated and analysed by immunoblotting to determine the abundance of AQP2 and its relative levels of tyrosine phosphorylation. No significant differences in the tyrosine phosphorylation levels of immunoprecipated-AQP2 were observed between groups. We can thus conclude that changes in the localization of AQP2 in the queen endometrium are not modulated by tyrosine phosphorylation.
Collapse
Affiliation(s)
- Lluis Ferré-Dolcet
- Unit of Animal Reproduction, Department of Animal Medicine and Surgery, Faculty of Veterinary Medicine, Autonomous University of Barcelona, Bellaterra, Spain
| | - Joan Enric Rodríguez-Gil
- Unit of Animal Reproduction, Department of Animal Medicine and Surgery, Faculty of Veterinary Medicine, Autonomous University of Barcelona, Bellaterra, Spain
| | - Marc Yeste
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain.,Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
| | - Teresa Rigau
- Unit of Animal Reproduction, Department of Animal Medicine and Surgery, Faculty of Veterinary Medicine, Autonomous University of Barcelona, Bellaterra, Spain
| | - Maria Montserrat Rivera Del Alamo
- Unit of Animal Reproduction, Department of Animal Medicine and Surgery, Faculty of Veterinary Medicine, Autonomous University of Barcelona, Bellaterra, Spain
| |
Collapse
|
205
|
Nishi M, Miyakawa K, Matsunaga S, Khatun H, Yamaoka Y, Watashi K, Sugiyama M, Kimura H, Wakita T, Ryo A. Prolyl Isomerase Pin1 Regulates the Stability of Hepatitis B Virus Core Protein. Front Cell Dev Biol 2020; 8:26. [PMID: 32083080 PMCID: PMC7005485 DOI: 10.3389/fcell.2020.00026] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/14/2020] [Indexed: 01/14/2023] Open
Abstract
The dynamic interplay between virus and host proteins is critical for establishing efficient viral replication and virus-induced pathogenesis. Phosphorylation-dependent prolyl isomerization by Pin1 provides a unique mechanism of molecular switching to control both protein function and stability. We demonstrate here that Pin1 binds and stabilizes hepatitis B virus core protein (HBc) in a phosphorylation-dependent manner, and promotes the efficient viral propagation. Phos-tag gel electrophoresis with various site-directed mutants of HBc revealed that Thr160 and Ser162 residues within the C terminal arginine-rich domain are phosphorylated concomitantly. GST pull-down assay and co-immunoprecipitation analysis demonstrated that Pin1 associated with phosphorylated HBc at the Thr160-Pro and Ser162-Pro motifs. Chemical or genetic inhibition of Pin1 significantly accelerated the rapid degradation of HBc via a lysosome-dependent pathway. Furthermore, we found that the pyruvate dehydrogenase phosphatase catalytic subunit 2 (PDP2) could dephosphorylate HBc at the Pin1-binding sites, thereby suppressing Pin1-mediated HBc stabilization. Our findings reveal an important regulatory mechanism of HBc stability catalyzed by Pin1 and may facilitate the development of new antiviral therapeutics targeting Pin1 function.
Collapse
Affiliation(s)
- Mayuko Nishi
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Kei Miyakawa
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Satoko Matsunaga
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Hajera Khatun
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Yutaro Yamaoka
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama, Japan.,Isehara Research Laboratory, Technology and Development Division, Kanto Chemical Co., Inc., Isehara, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masaya Sugiyama
- Genome Medical Sciences Project, National Center for Global Health and Medicine, Chiba, Japan
| | - Hirokazu Kimura
- Faculty of Health Sciences, School of Medical Technology, Gunma Paz University, Takasaki, Japan
| | - Takaji Wakita
- Genome Medical Sciences Project, National Center for Global Health and Medicine, Chiba, Japan
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama, Japan
| |
Collapse
|
206
|
Arora N, Raj A, Anjum F, Kaur R, Rawat SS, Kumar R, Tripathi S, Singh G, Prasad A. Unveiling Taenia solium kinome profile and its potential for new therapeutic targets. Expert Rev Proteomics 2020; 17:85-94. [DOI: 10.1080/14789450.2020.1719835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Naina Arora
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Anand Raj
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
- Department of Biotechnology, Motilal Nehru Institute of Technology, Allahabad, India
| | - Farhan Anjum
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Rimanpreet Kaur
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Suraj Singh Rawat
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Rajiv Kumar
- Department of Biotechnology, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India
| | - Shweta Tripathi
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Gagandeep Singh
- Department of Neurology, Dayanand Medical College, Ludhiana, India
| | - Amit Prasad
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| |
Collapse
|
207
|
Abstract
Adaptive mutations play an important role in molecular evolution. However, the frequency and nature of these mutations at the intramolecular level are poorly understood. To address this, we analyzed the impact of protein architecture on the rate of adaptive substitutions, aiming to understand how protein biophysics influences fitness and adaptation. Using Drosophila melanogaster and Arabidopsis thaliana population genomics data, we fitted models of distribution of fitness effects and estimated the rate of adaptive amino-acid substitutions both at the protein and amino-acid residue level. We performed a comprehensive analysis covering genome, gene, and protein structure, by exploring a multitude of factors with a plausible impact on the rate of adaptive evolution, such as intron number, protein length, secondary structure, relative solvent accessibility, intrinsic protein disorder, chaperone affinity, gene expression, protein function, and protein-protein interactions. We found that the relative solvent accessibility is a major determinant of adaptive evolution, with most adaptive mutations occurring at the surface of proteins. Moreover, we observe that the rate of adaptive substitutions differs between protein functional classes, with genes encoding for protein biosynthesis and degradation signaling exhibiting the fastest rates of protein adaptation. Overall, our results suggest that adaptive evolution in proteins is mainly driven by intermolecular interactions, with host-pathogen coevolution likely playing a major role.
Collapse
Affiliation(s)
- Ana Filipa Moutinho
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Fernanda Fontes Trancoso
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Julien Yann Dutheil
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Biology, Plön, Germany.,Unité Mixte de Recherche 5554 Institut des Sciences de l'Evolution, CNRS, IRD, EPHE, Université de Montpellier, Montpellier, France
| |
Collapse
|
208
|
Yan J, Long Y, Zhou T, Ren J, Li Q, Song G, Cui Z. Dynamic Phosphoproteome Profiling of Zebrafish Embryonic Fibroblasts during Cold Acclimation. Proteomics 2020; 20:e1900257. [PMID: 31826332 DOI: 10.1002/pmic.201900257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/24/2019] [Indexed: 11/09/2022]
Abstract
Temperature affects almost all aspects of the fish life. To cope with low temperature, fish have evolved the ability of cold acclimation for survival. However, intracellular signaling events underlying cold acclimation in fish remain largely unknown. Here, the formation of cold acclimation in zebrafish embryonic fibroblasts (ZF4) is monitored and the phosphorylation events during the process are investigated through a large-scale quantitative phosphoproteomic approach. In total, 11 474 phosphorylation sites are identified on 4066 proteins and quantified 5772 phosphosites on 2519 proteins. Serine, threonine, and tyrosine (Ser/Thr/Tyr) phosphorylation accounted for 85.5%, 13.3%, and 1.2% of total phosphosites, respectively. Among all phosphosites, 702 phosphosites on 510 proteins show differential regulation during cold acclimation of ZF4 cells. These phosphosites are divided into six clusters according to their dynamic changes during cold exposure. Kinase-substrate prediction reveals that mitogen-activated protein kinase (MAPK) among the kinase groups is predominantly responsible for phosphorylation of these phosphosites. The differentially regulated phosphoproteins are functionally associated with various cellular processes such as regulation of actin cytoskeleton and MAPK signaling pathway. These data enrich the database of protein phosphorylation sites in zebrafish and provide key clues for the elucidation of intracellular signaling networks during cold acclimation of fish.
Collapse
Affiliation(s)
- Junjun Yan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei, Wuhan, 430072, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yong Long
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei, Wuhan, 430072, China
| | - Tong Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei, Wuhan, 430072, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Ren
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei, Wuhan, 430072, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei, Wuhan, 430072, China
| | - Guili Song
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei, Wuhan, 430072, China
| | - Zongbin Cui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Hubei, Wuhan, 430072, China.,The Innovative Academy of Seed Design, Chinese Academy of Sciences, Hubei, Wuhan, 430072, China
| |
Collapse
|
209
|
N-Terminal Acetylation by NatB Is Required for the Shutoff Activity of Influenza A Virus PA-X. Cell Rep 2020; 24:851-860. [PMID: 30044982 DOI: 10.1016/j.celrep.2018.06.078] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 03/26/2018] [Accepted: 06/18/2018] [Indexed: 12/17/2022] Open
Abstract
N-terminal acetylation is a major posttranslational modification in eukaryotes catalyzed by N-terminal acetyltransferases (NATs), NatA through NatF. Although N-terminal acetylation modulates diverse protein functions, little is known about its roles in virus replication. We found that NatB, which comprises NAA20 and NAA25, is involved in the shutoff activity of influenza virus PA-X. The shutoff activity of PA-X was suppressed in NatB-deficient cells, and PA-X mutants that are not acetylated by NatB showed reduced shutoff activities. We also evaluated the importance of N-terminal acetylation of PA, because PA-X shares its N-terminal sequence with PA. Viral polymerase activity was reduced in NatB-deficient cells. Moreover, mutant PAs that are not acetylated by NatB lost their function in the viral polymerase complex. Taken together, our findings demonstrate that N-terminal acetylation is required for the shutoff activity of PA-X and for viral polymerase activity.
Collapse
|
210
|
Zapata-Carmona H, Soriano-Úbeda C, París-Oller E, Matás C. Periovulatory oviductal fluid decreases sperm protein kinase A activity, tyrosine phosphorylation, and in vitro fertilization in pig. Andrology 2020; 8:756-768. [PMID: 31872543 DOI: 10.1111/andr.12751] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 12/05/2019] [Accepted: 12/20/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Molecules from the female reproductive tract modulate capacitation and function of sperm cells in vivo. These molecules vary in a quantitative and qualitative manner throughout the estrous cycle. OBJECTIVES This work evaluates the effect of using various female reproductive fluids on capacitation and fertilization of pig spermatozoa in vitro. MATERIAL AND METHODS The effects of culturing spermatozoa in different fluids on the levels of sperm protein kinase A (pPKA), tyrosine phosphorylation, acrosome reaction, and in vitro fertilization (IVF) were evaluated. The fluids tested were as follows: oviductal fluid (OF) from five phases of the estrous cycle, namely early and late follicular (OF-EF, OF-LF), early and late luteal (OF-EL, OF-LL) and periovulatory (pOF), follicular fluid from medium-sized follicles, and secretions of cumulus-oocyte complexes (conditioned medium). RESULTS The pPKAs and tyrosine phosphorylation were decreased by OF-EF, OF-LF, OF-EL, and pOF but not by follicular fluid and conditioned medium. OF-EF, OF-LF, and pOF also decreased the sperm acrosome reaction. Moreover, the effect of pOF on pPKAs and tyrosine phosphorylation was reversible. In in vitro fertilization, OF-EF, OF-LF, OF-EL, and pOF reduced the percentage of penetrated oocytes, the mean number of spermatozoa per penetrated oocyte, and increased monospermy. CONCLUSION OF from follicular, early luteal, and periovulatory phases of the estrous cycle modulates the sperm protein phosphorylation as well as the acrosome reaction involved in capacitation and increases monospermic fertilization in in vitro fertilization. Our findings suggest that fluids from the female reproductive tract could be used as additives in porcine IVF systems to modulate sperm-oocyte interaction.
Collapse
Affiliation(s)
- Héctor Zapata-Carmona
- Department of Physiology, Faculty of Veterinary Science, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, Murcia, Spain.,Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Murcia, Spain.,Laboratorio de Biología de la Reproducción, Departamento Biomédico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Cristina Soriano-Úbeda
- Department of Physiology, Faculty of Veterinary Science, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, Murcia, Spain.,Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Murcia, Spain
| | - Evelyne París-Oller
- Department of Physiology, Faculty of Veterinary Science, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, Murcia, Spain.,Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Murcia, Spain
| | - Carmen Matás
- Department of Physiology, Faculty of Veterinary Science, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, Murcia, Spain.,Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Murcia, Spain
| |
Collapse
|
211
|
Benedetti AM, Gill DM, Tsang CW, Jones AM. Chemical Methods for N- and O-Sulfation of Small Molecules, Amino Acids and Peptides. Chembiochem 2020; 21:938-942. [PMID: 31692230 DOI: 10.1002/cbic.201900673] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Indexed: 11/06/2022]
Abstract
Sulfation of the amino acid residues of proteins is a significant post-translational modification, the functions of which are yet to be fully understood. Current sulfation methods are limited mainly to O-tyrosine (sY), which requires negatively charged species around the desired amino acid residue and a specific sulfotransferase enzyme. Alternatively, for solid-phase peptide synthesis, a de novo protected sY is required. Therefore, synthetic routes that go beyond O-sulfation are required. We have developed a novel route to N-sulfamation and can dial-in/out O-sulfation (without S-sulfurothiolation), mimicking the initiation step of the ping-pong sulfation mechanism identified in structural biology. This rapid, low-temperature and non-racemising method is applicable to a range of amines, amides, amino acids, and peptide sequences.
Collapse
Affiliation(s)
- Anna Mary Benedetti
- School of Pharmacy, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Daniel M Gill
- School of Pharmacy, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Chi W Tsang
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Alan M Jones
- School of Pharmacy, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|
212
|
Wang Y, Wang P, Xu J. Phosphorylation: A Fast Switch For Checkpoint Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:347-398. [PMID: 32185718 DOI: 10.1007/978-981-15-3266-5_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Checkpoint signaling involves a variety of upstream and downstream factors that participate in the regulation of checkpoint expression, activation, and degradation. During the process, phosphorylation plays a critical role. Phosphorylation is one of the most well-documented post-translational modifications of proteins. Of note, the importance of phosphorylation has been emphasized in aspects of cell activities, including proliferation, metabolism, and differentiation. Here we summarize how phosphorylation of specific molecules affects the immune activities with preference in tumor immunity. Of course, immune checkpoints are given extra attention in this book. There are many common pathways that are involved in signaling of different checkpoints. Some of them are integrated and presented as common activities in the early part of this chapter, especially those associated with PD-1/PD-L1 and CTLA-4, because investigations concerning them are particularly abundant and variant. Their distinct regulation is supplementarily discussed in their respective section. As for checkpoints that are so far not well explored, their related phosphorylation modulations are listed separately in the later part. We hope to provide a clear and systematic view of the phosphorylation-modulated immune signaling.
Collapse
Affiliation(s)
- Yiting Wang
- School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Wang
- Shanghai Tenth People's Hospital of Tongji University, School of Medicine, School of Life Sciences and Technology, Tongji University Cancer Center, Tongji University, Shanghai, 200092, China
| | - Jie Xu
- Institutes of Biomedical Sciences, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
213
|
Hasan R, Leo MD, Muralidharan P, Mata-Daboin A, Yin W, Bulley S, Fernandez-Peña C, MacKay CE, Jaggar JH. SUMO1 modification of PKD2 channels regulates arterial contractility. Proc Natl Acad Sci U S A 2019; 116:27095-27104. [PMID: 31822608 PMCID: PMC6936352 DOI: 10.1073/pnas.1917264116] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
PKD2 (polycystin-2, TRPP1) channels are expressed in a wide variety of cell types and can regulate functions, including cell division and contraction. Whether posttranslational modification of PKD2 modifies channel properties is unclear. Similarly uncertain are signaling mechanisms that regulate PKD2 channels in arterial smooth muscle cells (myocytes). Here, by studying inducible, cell-specific Pkd2 knockout mice, we discovered that PKD2 channels are modified by SUMO1 (small ubiquitin-like modifier 1) protein in myocytes of resistance-size arteries. At physiological intravascular pressures, PKD2 exists in approximately equal proportions as either nonsumoylated (PKD2) or triple SUMO1-modifed (SUMO-PKD2) proteins. SUMO-PKD2 recycles, whereas unmodified PKD2 is surface-resident. Intravascular pressure activates voltage-dependent Ca2+ influx that stimulates the return of internalized SUMO-PKD2 channels to the plasma membrane. In contrast, a reduction in intravascular pressure, membrane hyperpolarization, or inhibition of Ca2+ influx leads to lysosomal degradation of internalized SUMO-PKD2 protein, which reduces surface channel abundance. Through this sumoylation-dependent mechanism, intravascular pressure regulates the surface density of SUMO-PKD2-mediated Na+ currents (INa) in myocytes to control arterial contractility. We also demonstrate that intravascular pressure activates SUMO-PKD2, not PKD2, channels, as desumoylation leads to loss of INa activation in myocytes and vasodilation. In summary, this study reveals that PKD2 channels undergo posttranslational modification by SUMO1, which enables physiological regulation of their surface abundance and pressure-mediated activation in myocytes and thus control of arterial contractility.
Collapse
Affiliation(s)
- Raquibul Hasan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - M. Dennis Leo
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | | | - Alejandro Mata-Daboin
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Wen Yin
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Simon Bulley
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Carlos Fernandez-Peña
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Charles E. MacKay
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Jonathan H. Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| |
Collapse
|
214
|
The role of phosphorylation of MLF2 at serine 24 in BCR-ABL leukemogenesis. Cancer Gene Ther 2019; 27:98-107. [PMID: 31831854 DOI: 10.1038/s41417-019-0152-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 01/29/2023]
Abstract
Chronic myelogenous leukemia (CML) is a myeloproliferative disorder defined by the presence of the fusion gene BCR-ABL1 in primitive hematopoietic progenitors. The myeloid leukemia factors (MLFs) were identified in the fly and human, and are involved in acute leukemia and enhancing the myeloid factor; however, the function of MLF2 in CML is poorly understood. In this study, we demonstrated that MLF2 may play an oncogenic role in CML. The expression level of MLF2 was related to the proliferation, colony-formation ability, and sensitivity to imatinib in K562 cells. Moreover, phosphorylation at serine 24, detected through Phos-tag sodium dodecyl sulfate-polyacrylamide gel electrophoresis, was required to maintain the activity of MLF2 in CML. The effects of MLF2 overexpression on the colony-formation ability in vitro and mouse survival in vivo could be alleviated by point mutation of MLF2 at serine 24. These findings uncover the oncogenic role of MLF2 through phosphorylation at serine 24 and provide a novel therapeutic target in CML.
Collapse
|
215
|
García-Blanco N, Vázquez-Bolado A, Moreno S. Greatwall-Endosulfine: A Molecular Switch that Regulates PP2A/B55 Protein Phosphatase Activity in Dividing and Quiescent Cells. Int J Mol Sci 2019; 20:ijms20246228. [PMID: 31835586 PMCID: PMC6941129 DOI: 10.3390/ijms20246228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/02/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022] Open
Abstract
During the cell cycle, hundreds of proteins become phosphorylated and dephosphorylated, indicating that protein kinases and protein phosphatases play a central role in its regulation. It has been widely recognized that oscillation in cyclin-dependent kinase (CDK) activity promotes DNA replication, during S-phase, and chromosome segregation, during mitosis. Each CDK substrate phosphorylation status is defined by the balance between CDKs and CDK-counteracting phosphatases. In fission yeast and animal cells, PP2A/B55 is the main protein phosphatase that counteracts CDK activity. PP2A/B55 plays a key role in mitotic entry and mitotic exit, and it is regulated by the Greatwall-Endosulfine (ENSA) molecular switch that inactivates PP2A/B55 at the onset of mitosis, allowing maximal CDK activity at metaphase. The Greatwall-ENSA-PP2A/B55 pathway is highly conserved from yeast to animal cells. In yeasts, Greatwall is negatively regulated by nutrients through TORC1 and S6 kinase, and couples cell growth, regulated by TORC1, to cell cycle progression, driven by CDK activity. In animal cells, Greatwall is phosphorylated and activated by Cdk1 at G2/M, generating a bistable molecular switch that results in full activation of Cdk1/CyclinB. Here we review the current knowledge of the Greatwall-ENSA-PP2A/B55 pathway and discuss its role in cell cycle progression and as an integrator of nutritional cues.
Collapse
|
216
|
Wu Z, Jin Y, Chen B, Gugger MK, Wilkinson-Johnson CL, Tiambeng TN, Jin S, Ge Y. Comprehensive Characterization of the Recombinant Catalytic Subunit of cAMP-Dependent Protein Kinase by Top-Down Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2019; 30:2561-2570. [PMID: 31792770 PMCID: PMC6922056 DOI: 10.1007/s13361-019-02341-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/04/2019] [Accepted: 09/09/2019] [Indexed: 05/22/2023]
Abstract
Reversible phosphorylation plays critical roles in cell growth, division, and signal transduction. Kinases which catalyze the transfer of γ-phosphate groups of nucleotide triphosphates to their substrates are central to the regulation of protein phosphorylation and are therefore important therapeutic targets. Top-down mass spectrometry (MS) presents unique opportunities to study protein kinases owing to its capabilities in comprehensive characterization of proteoforms that arise from alternative splicing, sequence variations, and post-translational modifications. Here, for the first time, we developed a top-down MS method to characterize the catalytic subunit (C-subunit) of an important kinase, cAMP-dependent protein kinase (PKA). The recombinant PKA C-subunit was expressed in Escherichia coli and successfully purified via his-tag affinity purification. By intact mass analysis with high resolution and high accuracy, four different proteoforms of the affinity-purified PKA C-subunit were detected, and the most abundant proteoform was found containing seven phosphorylations with the removal of N-terminal methionine. Subsequently, the seven phosphorylation sites of the most abundant PKA C-subunit proteoform were characterized simultaneously using tandem MS methods. Four sites were unambiguously identified as Ser10, Ser11, Ser18, and Ser30, and the remaining phosphorylation sites were localized to Ser2/Ser3, Ser358/Thr368, and Thr[215-224]Tyr in the PKA C-subunit sequence with a 20mer 6xHis-tag added at the N-terminus. Interestingly, four of these seven phosphorylation sites were located at the 6xHis-tag. Furthermore, we have performed dephosphorylation reaction by Lambda protein phosphatase and showed that all phosphorylations of the recombinant PKA C-subunit phosphoproteoforms were removed by this phosphatase.
Collapse
Affiliation(s)
- Zhijie Wu
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Yutong Jin
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Bifan Chen
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Morgan K Gugger
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | | | - Timothy N Tiambeng
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Song Jin
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Human Proteomics Program, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
217
|
Zhou S, Zheng WJ, Liu BH, Zheng JC, Dong FS, Liu ZF, Wen ZY, Yang F, Wang HB, Xu ZS, Zhao H, Liu YW. Characterizing the Role of TaWRKY13 in Salt Tolerance. Int J Mol Sci 2019; 20:ijms20225712. [PMID: 31739570 PMCID: PMC6888956 DOI: 10.3390/ijms20225712] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 11/09/2019] [Accepted: 11/11/2019] [Indexed: 01/09/2023] Open
Abstract
The WRKY transcription factor superfamily is known to participate in plant growth and stress response. However, the role of this family in wheat (Triticum aestivum L.) is largely unknown. Here, a salt-induced gene TaWRKY13 was identified in an RNA-Seq data set from salt-treated wheat. The results of RT-qPCR analysis showed that TaWRKY13 was significantly induced in NaCl-treated wheat and reached an expression level of about 22-fold of the untreated wheat. Then, a further functional identification was performed in both Arabidopsis thaliana and Oryza sativa L. Subcellular localization analysis indicated that TaWRKY13 is a nuclear-localized protein. Moreover, various stress-related regulatory elements were predicted in the promoter. Expression pattern analysis revealed that TaWRKY13 can also be induced by polyethylene glycol (PEG), exogenous abscisic acid (ABA), and cold stress. After NaCl treatment, overexpressed Arabidopsis lines of TaWRKY13 have a longer root and a larger root surface area than the control (Columbia-0). Furthermore, TaWRKY13 overexpression rice lines exhibited salt tolerance compared with the control, as evidenced by increased proline (Pro) and decreased malondialdehyde (MDA) contents under salt treatment. The roots of overexpression lines were also more developed. These results demonstrate that TaWRKY13 plays a positive role in salt stress.
Collapse
Affiliation(s)
- Shuo Zhou
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
| | - Wei-Jun Zheng
- College of Agronomy, Northwest A&F University, Yangling 712100, China;
| | - Bao-Hua Liu
- Handan Academy of Agricultural Sciences, Handan 056001, China;
| | - Jia-Cheng Zheng
- College of Agronomy, Anhui Science and Technology University, Fengyang, Chuzhou 239000, China;
| | - Fu-Shuang Dong
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
| | | | - Zhi-Yu Wen
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
| | - Fan Yang
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
| | - Hai-Bo Wang
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
| | - Zhao-Shi Xu
- Institute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)/National Key Facility for Crop Gene Resources and Genetic Improvement, Key Laboratory of Biology and Genetic Improvement of Triticeae Crops, Ministry of Agriculture, Beijing 100081, China;
| | - He Zhao
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
- Correspondence: (H.Z.); (Y.-W.L.)
| | - Yong-Wei Liu
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences/Plant Genetic Engineering Center of Hebei Province, Shijiazhuang 050051, China; (S.Z.); (F.-S.D.); (Z.-Y.W.); (H.-B.W.)
- Correspondence: (H.Z.); (Y.-W.L.)
| |
Collapse
|
218
|
Fan K, Yuan S, Chen J, Chen Y, Li Z, Lin W, Zhang Y, Liu J, Lin W. Molecular evolution and lineage-specific expansion of the PP2C family in Zea mays. PLANTA 2019; 250:1521-1538. [PMID: 31346803 DOI: 10.1007/s00425-019-03243-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/16/2019] [Indexed: 05/19/2023]
Abstract
97 ZmPP2Cs were clustered into 10 subfamilies with biased subfamily evolution and lineage-specific expansion. Segmental duplication after the divergence of maize and sorghum might have led to primary expansion of ZmPP2Cs. The protein phosphatase 2C (PP2C) enzymes control many stress responses and developmental processes in plants. In Zea mays, a comprehensive understanding of the evolution and expansion of the PP2C family is still lacking. In the current study, 97 ZmPP2Cs were identified and clustered into 10 subfamilies. Through the analysis of the PP2C family in monocots, the ZmPP2C subfamilies displayed biased subfamily molecular evolution and lineage-specific expansion, as evidenced by their differing numbers of member genes, expansion and evolutionary rates, conserved subdomains, chromosomal distributions, expression levels, responsive-regulatory elements and regulatory networks. Moreover, while segmental duplication events have caused the primary expansion of the ZmPP2Cs, the majority of their diversification occurred following the additional whole-genome duplication that took place after the divergence of maize and sorghum (Sorghum bicolor). After this event, the PP2C subfamilies showed asymmetric evolutionary rates, with the D, F2 and H subfamily likely the most closely to resemble its ancestral subfamily's genes. These findings could provide novel insights into the molecular evolution and expansion of the PP2C family in maize, and lay the foundation for the functional analysis of these enzymes in maize and related monocots.
Collapse
Affiliation(s)
- Kai Fan
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Crop Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Crop Ecology and Molecular Physiology (Fujian Agriculture and Forestry University), Fujian Province University, Fuzhou, 35002, China
| | - Shuna Yuan
- Rubber Research Institute, Chinese Academy of Tropical Agricultural Sciences/Danzhou Investigation and Experiment Station of Tropical Crops, Ministry of Agriculture, Danzhou, 571737, China
| | - Jie Chen
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Crop Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Crop Ecology and Molecular Physiology (Fujian Agriculture and Forestry University), Fujian Province University, Fuzhou, 35002, China
| | - Yunrui Chen
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Crop Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Crop Ecology and Molecular Physiology (Fujian Agriculture and Forestry University), Fujian Province University, Fuzhou, 35002, China
| | - Zhaowei Li
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Crop Ecology and Molecular Physiology (Fujian Agriculture and Forestry University), Fujian Province University, Fuzhou, 35002, China
| | - Weiwei Lin
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Crop Ecology and Molecular Physiology (Fujian Agriculture and Forestry University), Fujian Province University, Fuzhou, 35002, China
| | - Yongqiang Zhang
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Crop Ecology and Molecular Physiology (Fujian Agriculture and Forestry University), Fujian Province University, Fuzhou, 35002, China
| | - Jianping Liu
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Crop Ecology and Molecular Physiology (Fujian Agriculture and Forestry University), Fujian Province University, Fuzhou, 35002, China
| | - Wenxiong Lin
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Crop Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
- Key Laboratory of Crop Ecology and Molecular Physiology (Fujian Agriculture and Forestry University), Fujian Province University, Fuzhou, 35002, China.
| |
Collapse
|
219
|
Siddiqui G, Proellochs NI, Cooke BM. Identification of essential exported
Plasmodium falciparum
protein kinases in malaria‐infected red blood cells. Br J Haematol 2019; 188:774-783. [DOI: 10.1111/bjh.16219] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/31/2019] [Indexed: 11/30/2022]
Affiliation(s)
- Ghizal Siddiqui
- Department of Microbiology Biomedicine Discovery Institute Monash University Clayton Victoria Australia
- Drug Delivery, Disposition and Dynamics Monash Institute of Pharmaceutical Sciences Monash University Parkville Victoria Australia
| | - Nicholas I. Proellochs
- Department of Microbiology Biomedicine Discovery Institute Monash University Clayton Victoria Australia
- Department of Medical Microbiology Radboud University Medical Center Nijmegen the Netherlands
| | - Brian M. Cooke
- Department of Microbiology Biomedicine Discovery Institute Monash University Clayton Victoria Australia
- Australian Institute of Tropical Health and Medicine James Cook University Cairns Queensland Australia
| |
Collapse
|
220
|
Vitrac H, Mallampalli VKPS, Dowhan W. Importance of phosphorylation/dephosphorylation cycles on lipid-dependent modulation of membrane protein topology by posttranslational phosphorylation. J Biol Chem 2019; 294:18853-18862. [PMID: 31645436 DOI: 10.1074/jbc.ra119.010785] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/10/2019] [Indexed: 12/29/2022] Open
Abstract
Posttranslational modifications of proteins, such as phosphorylation and dephosphorylation, play critical roles in cellular functions through diverse cell signaling pathways. Protein kinases and phosphatases have been described early on as key regulatory elements of the phosphorylated state of proteins. Tight spatial and temporal regulation of protein kinase and phosphatase activities has to be achieved in the cell to ensure accurate signal transduction. We demonstrated previously that phosphorylation of a membrane protein can lead to its topological rearrangement. Additionally, we found that both the rate and extent of topological rearrangement upon phosphorylation are lipid charge- and lipid environment-dependent. Here, using a model membrane protein (the bacterial lactose permease LacY reconstituted in proteoliposomes) and a combination of real-time measurements and steady-state assessments of protein topology, we established a set of experimental conditions to dissect the effects of phosphorylation and dephosphorylation of a membrane protein on its topological orientation. We also demonstrate that the phosphorylation-induced topological switch of a membrane protein can be reversed upon protein dephosphorylation, revealing a new regulatory role for phosphorylation/dephosphorylation cycles. Furthermore, we determined that the rate of topological rearrangement reversal is correlated with phosphatase activity and is influenced by the membrane's lipid composition, presenting new insights into the spatiotemporal control of the protein phosphorylation state. Together, our results highlight the importance of the compartmentalization of phosphorylation/dephosphorylation cycles in controlling membrane protein topology and, therefore, function, which are influenced by the local lipid environment of the membrane protein.
Collapse
Affiliation(s)
- Heidi Vitrac
- Department of Biochemistry and Molecular Biology and the Center for Membrane Biology, McGovern Medical School, University of Texas Houston, Texas 77030.
| | - Venkata K P S Mallampalli
- Department of Biochemistry and Molecular Biology and the Center for Membrane Biology, McGovern Medical School, University of Texas Houston, Texas 77030
| | - William Dowhan
- Department of Biochemistry and Molecular Biology and the Center for Membrane Biology, McGovern Medical School, University of Texas Houston, Texas 77030.
| |
Collapse
|
221
|
Sharma R, Sharma A, Kumar A, Jaganathan BG. Phospho-protein Analysis in Adherent Cells Using Flow Cytometry. Bio Protoc 2019; 9:e3395. [PMID: 33654896 DOI: 10.21769/bioprotoc.3395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 11/02/2022] Open
Abstract
Protein phosphorylation is one of the most important post-translational modifications, which acts as a reversible on or off switch for the activity of a large number of proteins. Analyzing the phosphorylation status of different proteins can reveal the alterations in the state of the cells in response to cellular damage, cancer and pharmaceutical drugs. Techniques such as mass spectrometry, radiolabeling, 2D-gel electrophoresis and western blotting are used to quantify protein phosphorylation. These assays can quantify phosphorylation in the bulk population of cells, however, flow cytometry can couple cell surface marker expression data with phosphorylation data to understand differential signaling in a sub-population within a heterogeneous population of cells. Our protocol describes the use of flow-cytometry for rapid and single cell-based quantification of intracellular phospho-protein with the help of anti-phospho protein specific antibody.
Collapse
Affiliation(s)
- Renu Sharma
- Stem Cells and Cancer Biology Group, Department of Biosciences and Bioengineering, Guwahati, India
| | - Amit Sharma
- Stem Cells and Cancer Biology Group, Department of Biosciences and Bioengineering, Guwahati, India
| | - Atul Kumar
- Stem Cells and Cancer Biology Group, Department of Biosciences and Bioengineering, Guwahati, India
| | - Bithiah Grace Jaganathan
- Stem Cells and Cancer Biology Group, Department of Biosciences and Bioengineering, Guwahati, India
| |
Collapse
|
222
|
Zhao X, Nedvetsky P, Stanchi F, Vion AC, Popp O, Zühlke K, Dittmar G, Klussmann E, Gerhardt H. Endothelial PKA activity regulates angiogenesis by limiting autophagy through phosphorylation of ATG16L1. eLife 2019; 8:e46380. [PMID: 31580256 PMCID: PMC6797479 DOI: 10.7554/elife.46380] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 10/01/2019] [Indexed: 12/15/2022] Open
Abstract
The cAMP-dependent protein kinase A (PKA) regulates various cellular functions in health and disease. In endothelial cells PKA activity promotes vessel maturation and limits tip cell formation. Here, we used a chemical genetic screen to identify endothelial-specific direct substrates of PKA in human umbilical vein endothelial cells (HUVEC) that may mediate these effects. Amongst several candidates, we identified ATG16L1, a regulator of autophagy, as novel target of PKA. Biochemical validation, mass spectrometry and peptide spot arrays revealed that PKA phosphorylates ATG16L1α at Ser268 and ATG16L1β at Ser269, driving phosphorylation-dependent degradation of ATG16L1 protein. Reducing PKA activity increased ATG16L1 protein levels and endothelial autophagy. Mouse in vivo genetics and pharmacological experiments demonstrated that autophagy inhibition partially rescues vascular hypersprouting caused by PKA deficiency. Together these results indicate that endothelial PKA activity mediates a critical switch from active sprouting to quiescence in part through phosphorylation of ATG16L1, which in turn reduces endothelial autophagy.
Collapse
Affiliation(s)
- Xiaocheng Zhao
- Vascular Patterning Laboratory, Center for Cancer BiologyVIBLeuvenBelgium
- Vascular Patterning Laboratory, Center for Cancer Biology, Department of OncologyVIBLeuvenBelgium
| | - Pavel Nedvetsky
- Vascular Patterning Laboratory, Center for Cancer BiologyVIBLeuvenBelgium
- Vascular Patterning Laboratory, Center for Cancer Biology, Department of OncologyVIBLeuvenBelgium
- Medical Cell Biology, Medical Clinic DUniversity Hospital MünsterMünsterGermany
| | - Fabio Stanchi
- Vascular Patterning Laboratory, Center for Cancer BiologyVIBLeuvenBelgium
- Vascular Patterning Laboratory, Center for Cancer Biology, Department of OncologyVIBLeuvenBelgium
| | - Anne-Clemence Vion
- Integrative Vascular Biology LabMax-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- INSERM UMR-970, Paris Cardiovascular Research CenterParis Descartes UniversityParisFrance
| | - Oliver Popp
- ProteomicsMax-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
| | - Kerstin Zühlke
- Anchored Signaling LabMax-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
| | - Gunnar Dittmar
- ProteomicsMax-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- CRP Santé · Department of OncologyLIH Luxembourg Institute of HealthLuxembourgLuxembourg
| | - Enno Klussmann
- Anchored Signaling LabMax-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- DZHK (German Center for Cardiovascular Research)BerlinGermany
| | - Holger Gerhardt
- Vascular Patterning Laboratory, Center for Cancer BiologyVIBLeuvenBelgium
- Vascular Patterning Laboratory, Center for Cancer Biology, Department of OncologyVIBLeuvenBelgium
- Integrative Vascular Biology LabMax-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- DZHK (German Center for Cardiovascular Research)BerlinGermany
- Berlin Institute of Health (BIH)BerlinGermany
| |
Collapse
|
223
|
Lu M, Ma L, Shan P, Liu A, Yu X, Jiang W, Wang X, Zhao X, Ye X, Wang T. DYRK1A aggravates β cell dysfunction and apoptosis by promoting the phosphorylation and degradation of IRS2. Exp Gerontol 2019; 125:110659. [PMID: 31306739 DOI: 10.1016/j.exger.2019.110659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 01/21/2023]
Abstract
In this study, we aimed to investigate the role of dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A (DYRK1A), which is one of the most important regulators of Alzheimer's disease development, in islet β cell dysfunction and apoptosis. We found significantly increased expression of DYRK1A in both the hippocampus and pancreatic islets of APPswe/PS1ΔE9 transgenic mice than in wild-type littermates. Furthermore, we observed that the overexpression of DYRK1A greatly aggravated β cell apoptosis. Most importantly, we found that DYRK1A directly interacted with insulin receptor substrate-2 (IRS2) and promoted IRS2 phosphorylation, leading to the proteasomal degradation of IRS2 and promotion of β cell dysfunction and apoptosis. These findings suggested that DYRK1A is a potential drug target in diabetes mellitus.
Collapse
Affiliation(s)
- Mei Lu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China; Shandong Provincial Key laboratory of Cardiovascular Proteomics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lin Ma
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China; Shandong Provincial Key laboratory of Cardiovascular Proteomics, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Peiyan Shan
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China; Shandong Provincial Key laboratory of Cardiovascular Proteomics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Aifen Liu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaolin Yu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wenjing Jiang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xinbang Wang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xinjing Zhao
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiang Ye
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Tan Wang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
224
|
Mullis BT, Hwang S, Lee LA, Iliuk A, Woolsey R, Quilici D, Wang Q. Automating Complex, Multistep Processes on a Single Robotic Platform to Generate Reproducible Phosphoproteomic Data. SLAS DISCOVERY 2019; 25:277-286. [PMID: 31556780 DOI: 10.1177/2472555219878152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mass spectrometry-based phosphoproteomics holds promise for advancing drug treatment and disease diagnosis; however, its clinical translation has thus far been limited. This is in part due to an unstandardized and segmented sample preparation process that involves cell lysis, protein digestion, peptide desalting, and phosphopeptide enrichment. Automating this entire sample preparation process will be key in facilitating standardization and clinical translation of phosphoproteomics. While peptide desalting and phosphopeptide enrichment steps have been individually automated, integrating these two extractions and, further, the entire process requires more advanced robotic platforms as well as automation-friendly extraction tools. Here we describe a fully automated peptide desalting and phosphopeptide enrichment method using IMCStips on a Hamilton STAR. Using our established automated method, we identified more than 10,000 phosphopeptides from 200 µg of HCT116 cell lysate without fractionation with >85% phosphopeptide specificities. Compared with titania-based Spin Tip products, the automated IMCStips-based method gave 50% higher phosphopeptide identifications. The method reproducibility was further assessed using multiple reaction monitoring (MRM) to show >50% phosphopeptide recoveries after the automated phosphopeptide extraction with coefficients of variation (CVs) of <20% over a 3-week period. The established automated method is a step toward standardization of the sample preparation of phosphopeptide samples and could be further expanded upon to create a fully automated "cells to phosphopeptides" method.
Collapse
Affiliation(s)
- B Todd Mullis
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Sunil Hwang
- UNC Nutrition Research Institute, Kannapolis, NC, USA
| | - L Andrew Lee
- Integrated Micro-Chromatography Systems, Inc., Irmo, SC, USA
| | - Anton Iliuk
- Tymora Analytical Operations, West Lafayette, IN, USA
| | - Rebekah Woolsey
- Mick Hitchcock, Ph.D. Nevada Proteomics Center, Reno, NV, USA
| | - David Quilici
- Mick Hitchcock, Ph.D. Nevada Proteomics Center, Reno, NV, USA
| | - Qian Wang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
225
|
Iida M, Sahashi K, Kondo N, Nakatsuji H, Tohnai G, Tsutsumi Y, Noda S, Murakami A, Onodera K, Okada Y, Nakatochi M, Tsukagoshi Okabe Y, Shimizu S, Mizuno M, Adachi H, Okano H, Sobue G, Katsuno M. Src inhibition attenuates polyglutamine-mediated neuromuscular degeneration in spinal and bulbar muscular atrophy. Nat Commun 2019; 10:4262. [PMID: 31537808 PMCID: PMC6753158 DOI: 10.1038/s41467-019-12282-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 08/29/2019] [Indexed: 12/12/2022] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) is a neuromuscular disease caused by an expanded CAG repeat in the androgen receptor (AR) gene. Here, we perform a comprehensive analysis of signaling pathways in a mouse model of SBMA (AR-97Q mice) utilizing a phosphoprotein assay. We measure the levels of 17 phosphorylated proteins in spinal cord and skeletal muscle of AR-97Q mice at three stages. The level of phosphorylated Src (p-Src) is markedly increased in the spinal cords and skeletal muscles of AR-97Q mice prior to the onset. Intraperitoneal administration of a Src kinase inhibitor improves the behavioral and histopathological phenotypes of the transgenic mice. We identify p130Cas as an effector molecule of Src and show that the phosphorylated p130Cas is elevated in murine and cellular models of SBMA. These results suggest that Src kinase inhibition is a potential therapy for SBMA.
Collapse
Affiliation(s)
- Madoka Iida
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8550, Japan
- Japan Society for the Promotion of Science, 5-3-1, Kojimachi, Chiyoda-ku, Tokyo, 102-0083, Japan
| | - Kentaro Sahashi
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8550, Japan
| | - Naohide Kondo
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8550, Japan
| | - Hideaki Nakatsuji
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8550, Japan
| | - Genki Tohnai
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8550, Japan
| | - Yutaka Tsutsumi
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8550, Japan
| | - Seiya Noda
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8550, Japan
- Department of Neurology, National Hospital Organization Suzuka National Hospital, 3-2-1, Kasado, Suzuka city, Mie, 513-8501, Japan
| | - Ayuka Murakami
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8550, Japan
- Department of Neurology, National Hospital Organization Suzuka National Hospital, 3-2-1, Kasado, Suzuka city, Mie, 513-8501, Japan
| | - Kazunari Onodera
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8550, Japan
- Department of Neurology, Aichi Medical University School of Medicine, 1, Karimata, Yazako, Nagakute-city, Aichi, 480-1195, Japan
| | - Yohei Okada
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8550, Japan
- Department of Neurology, Aichi Medical University School of Medicine, 1, Karimata, Yazako, Nagakute-city, Aichi, 480-1195, Japan
- Department of Physiology, Keio University School of Medicine, 35, Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masahiro Nakatochi
- Department of Nursing, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya city, Aichi, 461-8673, Japan
| | - Yuka Tsukagoshi Okabe
- Department of Advanced Medicine, Nagoya University Hospital, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8560, Japan
| | - Shinobu Shimizu
- Department of Advanced Medicine, Nagoya University Hospital, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8560, Japan
| | - Masaaki Mizuno
- Department of Advanced Medicine, Nagoya University Hospital, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8560, Japan
| | - Hiroaki Adachi
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, 1-1, Iseigaoka, Yahatanichi-ku, Kitakyushu-city, Fukuoka, 807-0804, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35, Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Gen Sobue
- Brain and Mind Research Center, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8550, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya city, Aichi, 466-8550, Japan.
| |
Collapse
|
226
|
Abstract
Castration-resistant prostate cancer (CRPC) remains incurable despite the approval of several new treatments. Identification of new biomarkers and therapeutic targets to enable personalization of CRPC therapy, with the aim of maximizing therapeutic responses and minimizing toxicity in patients, is urgently needed. Prostate cancer progression and therapeutic resistance are frequently driven by aberrantly activated kinase signalling pathways that are amenable to pharmacological inhibition. Personalized phosphoproteomics, which enables the analysis of signalling networks in individual tumours, is a promising approach to advance personalized therapy by discovering biomarkers of pathway activity and clinically actionable targets. Several technologies for global and targeted phosphoproteomic analysis exist, each with its own strengths and shortcomings. Global discovery phosphoproteomics is predominantly conducted using liquid chromatography-tandem mass spectrometry coupled with data-dependent or data-independent acquisition technologies. Multiplexed targeted phosphoproteomics can be divided into platforms based on mass spectrometry or antibodies, including selected or parallel reaction monitoring and triggered by offset, multiplexed, accurate mass, high-resolution, absolute quantification (known as TOMAHAQ) or forward-phase or reverse-phase protein arrays, respectively. Several obstacles still need to be overcome before the full potential of phosphoproteomics can be realized in routine clinical practice, but a future phosphoproteomics-centric trans-omic profiling approach should enable optimized personalized CRPC management through improved biomarkers and targeted treatments.
Collapse
|
227
|
Kim VY, Batty A, Li J, Kirk SG, Crowell SA, Jin Y, Tang J, Zhang J, Rogers LK, Deng HX, Nelin LD, Liu Y. Glutathione Reductase Promotes Fungal Clearance and Suppresses Inflammation during Systemic Candida albicans Infection in Mice. THE JOURNAL OF IMMUNOLOGY 2019; 203:2239-2251. [PMID: 31501257 DOI: 10.4049/jimmunol.1701686] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 08/07/2019] [Indexed: 01/16/2023]
Abstract
Glutathione reductase (Gsr) catalyzes the reduction of glutathione disulfide to glutathione, which plays an important role in redox regulation. We have previously shown that Gsr facilitates neutrophil bactericidal activities and is pivotal for host defense against bacterial pathogens. However, it is unclear whether Gsr is required for immune defense against fungal pathogens. It is also unclear whether Gsr plays a role in immunological functions outside of neutrophils during immune defense. In this study, we report that Gsr-/- mice exhibited markedly increased susceptibility to Candida albicans challenge. Upon C. albicans infection, Gsr-/- mice exhibited dramatically increased fungal burden in the kidneys, cytokine and chemokine storm, striking neutrophil infiltration, histological abnormalities in both the kidneys and heart, and substantially elevated mortality. Large fungal foci surrounded by massive numbers of neutrophils were detected outside of the glomeruli in the kidneys of Gsr -/- mice but were not found in wild-type mice. Examination of the neutrophils and macrophages of Gsr-/- mice revealed several defects. Gsr -/- neutrophils exhibited compromised phagocytosis, attenuated respiratory burst, and impaired fungicidal activity in vitro. Moreover, upon C. albicans stimulation, Gsr -/- macrophages produced increased levels of inflammatory cytokines and exhibited elevated p38 and JNK activities, at least in part, because of lower MAPK phosphatase (Mkp)-1 activity and greater Syk activity. Thus, Gsr-mediated redox regulation is crucial for fungal clearance by neutrophils and the proper control of the inflammatory response by macrophages during host defense against fungal challenge.
Collapse
Affiliation(s)
- Victoria Y Kim
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215
| | - Abel Batty
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215
| | - Jinhui Li
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215
| | - Sean G Kirk
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215
| | - Sara A Crowell
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215
| | - Yi Jin
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215
| | - Juan Tang
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210
| | - Jian Zhang
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Lynette K Rogers
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43205; and
| | - Han-Xiang Deng
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Leif D Nelin
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43205; and
| | - Yusen Liu
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215; .,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43205; and
| |
Collapse
|
228
|
Ding T, Zhou Y, Long R, Chen C, Zhao J, Cui P, Guo M, Liang G, Xu L. DUSP8 phosphatase: structure, functions, expression regulation and the role in human diseases. Cell Biosci 2019; 9:70. [PMID: 31467668 PMCID: PMC6712826 DOI: 10.1186/s13578-019-0329-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/12/2019] [Indexed: 12/28/2022] Open
Abstract
Dual-specificity phosphatases (DUSPs) are a subset of protein tyrosine phosphatases (PTPs), many of which dephosphorylate the residues of phosphor-serine/threonine and phosphor-tyrosine on mitogen-activated protein kinases (MAPKs), and hence are also referred to as MAPK phosphatases (MKPs). Homologue of Vaccinia virus H1 phosphatase gene clone 5 (HVH-5), also known as DUSP8, is a unique member of the DUSPs family of phosphatases. Accumulating evidence has shown that DUSP8 plays an important role in phosphorylation-mediated signal transduction of MAPK signaling ranging from cell oxidative stress response, cell apoptosis and various human diseases. It is generally believed that DUSP8 exhibits significant dephosphorylation activity against JNK, however, with the deepening of research, plenty of new literature reports that DUSP8 also has effective dephosphorylation activity on p38 MAPK and ERKs, successfully affects the transduction of MAPKs pathway, indicating that DUSP8 presents a unknown diversity of DUSPs family on distinct corresponding dephosphorylated substrates in different biological events. Therefore, the in-depth study of DUSP8 not only throws a new light on the multi-biological function of DUSPs, but also is much valuable for the reveal of complex pathobiology of clinical diseases. In this review, we provide a detail overview of DUSP8 phosphatase structure, biological function and expression regulation, as well as its role in related clinical human diseases, which might be help for the understanding of biological function of DUSP8 and the development of prevention, diagnosis and therapeutics in related human diseases.
Collapse
Affiliation(s)
- Tao Ding
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China.,2Department of Immunology, Zunyi Medical University, Zunyi, 563000 Guizhou China
| | - Ya Zhou
- 3Department of Medical Physics, Zunyi Medical University, Zunyi, 563000 Guizhou China
| | - Runying Long
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China.,2Department of Immunology, Zunyi Medical University, Zunyi, 563000 Guizhou China
| | - Chao Chen
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China
| | - Panpan Cui
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China
| | - Mengmeng Guo
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China
| | - Guiyou Liang
- 4Department of Cardiovascular Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China.,5Department of Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000 Guizhou China
| | - Lin Xu
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000 Guizhou China.,2Department of Immunology, Zunyi Medical University, Zunyi, 563000 Guizhou China
| |
Collapse
|
229
|
Das S, Bhattacharya B, Das B, Sinha B, Jamatia T, Paul K. Etiologic Role of Kinases in the Progression of Human Cancers and Its Targeting Strategies. Indian J Surg Oncol 2019; 12:34-45. [PMID: 33994726 DOI: 10.1007/s13193-019-00972-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 08/07/2019] [Indexed: 11/30/2022] Open
Abstract
Cancer is one of the dominant causes of death worldwide while lifelong prognosis is still inauspicious. The maturation of the cancer is seen as a process of transformation of a healthy cell into a tumor-sensitive cell, which is held entirely at the cellular, molecular, and genetic levels of the organism. Tyrosine kinases can play a major, etiologic role in the inception of malignancy and devote to the uncontrolled proliferation of cancerous cells and the progression of a tumor as well as the development of metastatic disease. Angiogenesis and oncogene activation are the major event in cell proliferation. The growth of a tumor and metastasis are fully depending on angiogenesis and lymphangiogenesis triggered by chemical signals from tumor cells in a phase of rapid growth. Tyrosine kinase inhibitors are compounds that inhibit tyrosine kinases and effective in targeting angiogenesis and blocking the signaling pathways of oncogenes. Small molecule tyrosine kinase inhibitors like afatinib, erlotinib, crizotinib, gefitinib, and cetuximab are shown to a selective cut off tactic toward the constitutive activation of an oncogene in tumor cells, and thus contemplated as promising therapeutic approaches for the diagnosis of cancer and malignancies.
Collapse
Affiliation(s)
- Sanjoy Das
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
| | - Bireswar Bhattacharya
- Regional Institute of Pharmaceutical Science and Technology, Agartala, Tripura 799005 India
| | - Biplajit Das
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
| | - Bibek Sinha
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
| | - Taison Jamatia
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
| | - Kishan Paul
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
| |
Collapse
|
230
|
Luan Z, Zhao L, Liu C, Song W, He P, Zhang X. Detection of casein kinase II by aggregation-induced emission. Talanta 2019; 201:450-454. [PMID: 31122448 DOI: 10.1016/j.talanta.2019.04.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/12/2019] [Accepted: 04/14/2019] [Indexed: 10/27/2022]
Abstract
A novel aggregation-induced emission (AIE) probe comprised of a hydrophilic protein kinase specific peptide and a hydrophobic tetraphenylethene (TPE) unit was synthesized through click reaction. The prepared TPE-peptide probe could be completely degraded by carboxypeptidase Y (CPY) to release hydrophobic TPE part, which aggregated in buffer solution and showed strong TPE emission. In the presence of casein kinase (CKII), the phosphorylation of peptide prevented the complete degradation by CPY producing the nonemissive probe. Thus, the developed probe can be used to detect CKII homogeneously and conveniently. This detection process can be finished within 1.5 h with high sensitivity (0.05 mU/μL) and good selectivity. The developed method can also be used to screen protein kinase inhibitor even in a complex biological system.
Collapse
Affiliation(s)
- Zhenzhu Luan
- Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, Key Laboratory of Biochemical Analysis, Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Li Zhao
- Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, Key Laboratory of Biochemical Analysis, Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Chao Liu
- Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, Key Laboratory of Biochemical Analysis, Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Weiling Song
- Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, Key Laboratory of Biochemical Analysis, Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Peng He
- Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, Key Laboratory of Biochemical Analysis, Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Xiaoru Zhang
- Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, Key Laboratory of Biochemical Analysis, Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China.
| |
Collapse
|
231
|
Li H, Guan Y. Machine learning empowers phosphoproteome prediction in cancers. Bioinformatics 2019; 36:859-864. [PMID: 31410451 PMCID: PMC7868059 DOI: 10.1093/bioinformatics/btz639] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/25/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023] Open
Abstract
MOTIVATION Reversible protein phosphorylation is an essential post-translational modification regulating protein functions and signaling pathways in many cellular processes. Aberrant activation of signaling pathways often contributes to cancer development and progression. The mass spectrometry-based phosphoproteomics technique is a powerful tool to investigate the site-level phosphorylation of the proteome in a global fashion, paving the way for understanding the regulatory mechanisms underlying cancers. However, this approach is time-consuming and requires expensive instruments, specialized expertise and a large amount of starting material. An alternative in silico approach is predicting the phosphoproteomic profiles of cancer patients from the available proteomic, transcriptomic and genomic data. RESULTS Here, we present a winning algorithm in the 2017 NCI-CPTAC DREAM Proteogenomics Challenge for predicting phosphorylation levels of the proteome across cancer patients. We integrate four components into our algorithm, including (i) baseline correlations between protein and phosphoprotein abundances, (ii) universal protein-protein interactions, (iii) shareable regulatory information across cancer tissues and (iv) associations among multi-phosphorylation sites of the same protein. When tested on a large held-out testing dataset of 108 breast and 62 ovarian cancer samples, our method ranked first in both cancer tissues, demonstrating its robustness and generalization ability. AVAILABILITY AND IMPLEMENTATION Our code and reproducible results are freely available on GitHub: https://github.com/GuanLab/phosphoproteome_prediction. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Hongyang Li
- To whom correspondence should be addressed. or
| | | |
Collapse
|
232
|
Przychodzen P, Kuban-Jankowska A, Wyszkowska R, Barone G, Bosco GL, Celso FL, Kamm A, Daca A, Kostrzewa T, Gorska-Ponikowska M. PTP1B phosphatase as a novel target of oleuropein activity in MCF-7 breast cancer model. Toxicol In Vitro 2019; 61:104624. [PMID: 31419504 DOI: 10.1016/j.tiv.2019.104624] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/30/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023]
Abstract
Phosphatase PTP1B has become a therapeutic target for the treatment of type 2-diabetes, whereas recent studies have revealed that PTP1B plays a pivotal role in pathophysiology and development of breast cancer. Oleuropein is a natural, phenolic compound with anticancer activity. The aim of this study was to address the question whether PTP1B constitutes a target for oleuropein in breast cancer MCF-7 cells. The cellular MCF-7 breast cancer model was used in the study. The experiments were performed using cellular viability tests, Elisa assays, immunoprecipitation, flow cytometry analyses and computer modelling. Herein, we evidenced that the reduced activity of phosphatase PTP1B after treatment with oleuropein is strictly correlated with decreased MCF-7 cellular viability and cell cycle arrest. These results provide new insight into further research on oleuropein and possible role of the compound in adjuvant treatment of breast cancer.
Collapse
Affiliation(s)
- Paulina Przychodzen
- Department of Medical Chemistry, Medical University of Gdansk, Gdansk, Poland
| | | | - Roksana Wyszkowska
- Department of Medical Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Giampaolo Barone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Giosuè Lo Bosco
- Department of Mathematics and Computer Science, University of Palermo, Palermo, Italy; The Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
| | - Fabrizio Lo Celso
- Department of Physics and Chemistry 'Emilio Segrè', University of Palermo, Palermo, Italy
| | - Anna Kamm
- Department of Medical Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Agnieszka Daca
- Department of Pathology and Experimental Rheumatology, Medical University of Gdansk, Gdansk, Poland
| | - Tomasz Kostrzewa
- Department of Medical Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Magdalena Gorska-Ponikowska
- Department of Medical Chemistry, Medical University of Gdansk, Gdansk, Poland; Institute of Biomaterials and Biomolecular Systems, Department of Biophysics, University of Stuttgart, Stuttgart, Germany; The Euro-Mediterranean Institute of Science and Technology, Palermo, Italy.
| |
Collapse
|
233
|
Arshad OA, Danna V, Petyuk VA, Piehowski PD, Liu T, Rodland KD, McDermott JE. An Integrative Analysis of Tumor Proteomic and Phosphoproteomic Profiles to Examine the Relationships Between Kinase Activity and Phosphorylation. Mol Cell Proteomics 2019; 18:S26-S36. [PMID: 31227600 PMCID: PMC6692771 DOI: 10.1074/mcp.ra119.001540] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/18/2019] [Indexed: 12/18/2022] Open
Abstract
Phosphorylation of proteins is a key way cells regulate function, both at the individual protein level and at the level of signaling pathways. Kinases are responsible for phosphorylation of substrates, generally on serine, threonine, or tyrosine residues. Though particular sequence patterns can be identified that dictate whether a residue will be phosphorylated by a specific kinase, these patterns are not highly predictive of phosphorylation. The availability of large scale proteomic and phosphoproteomic data sets generated using mass-spectrometry-based approaches provides an opportunity to study the important relationship between kinase activity, substrate specificity, and phosphorylation. In this study, we analyze relationships between protein abundance and phosphopeptide abundance across more than 150 tumor samples and show that phosphorylation at specific phosphosites is not well correlated with overall kinase abundance. However, individual kinases show a clear and statistically significant difference in correlation among known phosphosite targets for that kinase and randomly selected phosphosites. We further investigate relationships between phosphorylation of known activating or inhibitory sites on kinases and phosphorylation of their target phosphosites. Combined with motif-based analysis, this approach can predict novel kinase targets and show which subsets of a kinase's target repertoire are specifically active in one condition versus another.
Collapse
Affiliation(s)
- Osama A Arshad
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352
| | - Vincent Danna
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352
| | - Paul D Piehowski
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352
| | - Tao Liu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352
| | - Karin D Rodland
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352; School of Medicine, Oregon Health & Sciences University, Portland, OR 97239
| | - Jason E McDermott
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352; School of Medicine, Oregon Health & Sciences University, Portland, OR 97239.
| |
Collapse
|
234
|
Kelley AR, Bach SB, Perry G. Analysis of post-translational modifications in Alzheimer's disease by mass spectrometry. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2040-2047. [DOI: 10.1016/j.bbadis.2018.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/19/2018] [Accepted: 11/04/2018] [Indexed: 01/09/2023]
|
235
|
Gil J, Betancourt LH, Pla I, Sanchez A, Appelqvist R, Miliotis T, Kuras M, Oskolas H, Kim Y, Horvath Z, Eriksson J, Berge E, Burestedt E, Jönsson G, Baldetorp B, Ingvar C, Olsson H, Lundgren L, Horvatovich P, Murillo JR, Sugihara Y, Welinder C, Wieslander E, Lee B, Lindberg H, Pawłowski K, Kwon HJ, Doma V, Timar J, Karpati S, Szasz AM, Németh IB, Nishimura T, Corthals G, Rezeli M, Knudsen B, Malm J, Marko-Varga G. Clinical protein science in translational medicine targeting malignant melanoma. Cell Biol Toxicol 2019; 35:293-332. [PMID: 30900145 PMCID: PMC6757020 DOI: 10.1007/s10565-019-09468-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/13/2019] [Indexed: 02/06/2023]
Abstract
Melanoma of the skin is the sixth most common type of cancer in Europe and accounts for 3.4% of all diagnosed cancers. More alarming is the degree of recurrence that occurs with approximately 20% of patients lethally relapsing following treatment. Malignant melanoma is a highly aggressive skin cancer and metastases rapidly extend to the regional lymph nodes (stage 3) and to distal organs (stage 4). Targeted oncotherapy is one of the standard treatment for progressive stage 4 melanoma, and BRAF inhibitors (e.g. vemurafenib, dabrafenib) combined with MEK inhibitor (e.g. trametinib) can effectively counter BRAFV600E-mutated melanomas. Compared to conventional chemotherapy, targeted BRAFV600E inhibition achieves a significantly higher response rate. After a period of cancer control, however, most responsive patients develop resistance to the therapy and lethal progression. The many underlying factors potentially causing resistance to BRAF inhibitors have been extensively studied. Nevertheless, the remaining unsolved clinical questions necessitate alternative research approaches to address the molecular mechanisms underlying metastatic and treatment-resistant melanoma. In broader terms, proteomics can address clinical questions far beyond the reach of genomics, by measuring, i.e. the relative abundance of protein products, post-translational modifications (PTMs), protein localisation, turnover, protein interactions and protein function. More specifically, proteomic analysis of body fluids and tissues in a given medical and clinical setting can aid in the identification of cancer biomarkers and novel therapeutic targets. Achieving this goal requires the development of a robust and reproducible clinical proteomic platform that encompasses automated biobanking of patient samples, tissue sectioning and histological examination, efficient protein extraction, enzymatic digestion, mass spectrometry-based quantitative protein analysis by label-free or labelling technologies and/or enrichment of peptides with specific PTMs. By combining data from, e.g. phosphoproteomics and acetylomics, the protein expression profiles of different melanoma stages can provide a solid framework for understanding the biology and progression of the disease. When complemented by proteogenomics, customised protein sequence databases generated from patient-specific genomic and transcriptomic data aid in interpreting clinical proteomic biomarker data to provide a deeper and more comprehensive molecular characterisation of cellular functions underlying disease progression. In parallel to a streamlined, patient-centric, clinical proteomic pipeline, mass spectrometry-based imaging can aid in interrogating the spatial distribution of drugs and drug metabolites within tissues at single-cell resolution. These developments are an important advancement in studying drug action and efficacy in vivo and will aid in the development of more effective and safer strategies for the treatment of melanoma. A collaborative effort of gargantuan proportions between academia and healthcare professionals has led to the initiation, establishment and development of a cutting-edge cancer research centre with a specialisation in melanoma and lung cancer. The primary research focus of the European Cancer Moonshot Lund Center is to understand the impact that drugs have on cancer at an individualised and personalised level. Simultaneously, the centre increases awareness of the relentless battle against cancer and attracts global interest in the exceptional research performed at the centre.
Collapse
Affiliation(s)
- Jeovanis Gil
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden.
| | - Lazaro Hiram Betancourt
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden.
| | - Indira Pla
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 205 02, Malmö, Sweden
| | - Aniel Sanchez
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 205 02, Malmö, Sweden
| | - Roger Appelqvist
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
| | - Tasso Miliotis
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
- Translational Science, Cardiovascular Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Magdalena Kuras
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
| | - Henriette Oskolas
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
| | - Yonghyo Kim
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
| | - Zsolt Horvath
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
| | - Jonatan Eriksson
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
| | - Ethan Berge
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
| | - Elisabeth Burestedt
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
| | - Göran Jönsson
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, 221 85, Lund, Sweden
| | - Bo Baldetorp
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, 221 85, Lund, Sweden
| | - Christian Ingvar
- Department of Surgery, Clinical Sciences, Lund University, SUS, Lund, Sweden
| | - Håkan Olsson
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, 221 85, Lund, Sweden
| | - Lotta Lundgren
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, 221 85, Lund, Sweden
- Department of Haematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Peter Horvatovich
- Department of Analytical Biochemistry, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Jimmy Rodriguez Murillo
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
| | - Yutaka Sugihara
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
| | - Charlotte Welinder
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, 221 85, Lund, Sweden
| | - Elisabet Wieslander
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, 221 85, Lund, Sweden
| | - Boram Lee
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
| | - Henrik Lindberg
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
| | - Krzysztof Pawłowski
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
- Department of Experimental Design and Bioinformatics, Faculty of Agriculture and Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Ho Jeong Kwon
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
- Chemical Genomics Global Research Lab, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Viktoria Doma
- Second Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Jozsef Timar
- Second Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Sarolta Karpati
- Department of Dermatology, Semmelweis University, Budapest, Hungary
| | - A Marcell Szasz
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, 221 85, Lund, Sweden
- Cancer Center, Semmelweis University, Budapest, 1083, Hungary
- MTA-TTK Momentum Oncology Biomarker Research Group, Hungarian Academy of Sciences, Budapest, 1117, Hungary
| | - István Balázs Németh
- Department of Dermatology and Allergology, University of Szeged, Szeged, H-6720, Hungary
| | - Toshihide Nishimura
- Clinical Translational Medicine Informatics, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
- Department of Surgery, Tokyo Medical University, 6-7-1 Nishishinjiku Shinjiku-ku, Tokyo, Japan
| | - Garry Corthals
- Van't Hoff Institute of Molecular Sciences, 1090 GS, Amsterdam, The Netherlands
| | - Melinda Rezeli
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
| | - Beatrice Knudsen
- Biomedical Sciences and Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Johan Malm
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 205 02, Malmö, Sweden
| | - György Marko-Varga
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84, Lund, Sweden
- Chemical Genomics Global Research Lab, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
- Department of Surgery, Tokyo Medical University, 6-7-1 Nishishinjiku Shinjiku-ku, Tokyo, Japan
| |
Collapse
|
236
|
Travers T, Kanagy WK, Mansbach RA, Jhamba E, Cleyrat C, Goldstein B, Lidke DS, Wilson BS, Gnanakaran S. Combinatorial diversity of Syk recruitment driven by its multivalent engagement with FcεRIγ. Mol Biol Cell 2019; 30:2331-2347. [PMID: 31216232 PMCID: PMC6743456 DOI: 10.1091/mbc.e18-11-0722] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 05/17/2019] [Accepted: 06/10/2019] [Indexed: 12/14/2022] Open
Abstract
Syk/Zap70 family kinases are essential for signaling via multichain immune-recognition receptors such as tetrameric (αβγ2) FcεRI. Syk activation is generally attributed to cis binding of its tandem SH2 domains to dual phosphotyrosines within FcεRIγ-ITAMs (immunoreceptor tyrosine-based activation motifs). However, the mechanistic details of Syk docking on γ homodimers are unresolved. Here, we estimate that multivalent interactions for WT Syk improve cis-oriented binding by three orders of magnitude. We applied molecular dynamics (MD), hybrid MD/worm-like chain polymer modeling, and live cell imaging to evaluate relative binding and signaling output for all possible cis and trans Syk-FcεRIγ configurations. Syk binding is likely modulated during signaling by autophosphorylation on Y130 in interdomain A, since a Y130E phosphomimetic form of Syk is predicted to lead to reduced helicity of interdomain A and alter Syk's bias for cis binding. Experiments in reconstituted γ-KO cells, whose γ subunits are linked by disulfide bonds, as well as in cells expressing monomeric ITAM or hemITAM γ-chimeras, support model predictions that short distances between γ ITAM pairs are required for trans docking. We propose that the full range of docking configurations improves signaling efficiency by expanding the combinatorial possibilities for Syk recruitment, particularly under conditions of incomplete ITAM phosphorylation.
Collapse
Affiliation(s)
- Timothy Travers
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545
- Center for Nonlinear Studies, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - William K. Kanagy
- Department of Pathology, University of New Mexico, Albuquerque, NM 87131
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131
| | - Rachael A. Mansbach
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Elton Jhamba
- Department of Pathology, University of New Mexico, Albuquerque, NM 87131
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131
| | - Cedric Cleyrat
- Department of Pathology, University of New Mexico, Albuquerque, NM 87131
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131
| | - Byron Goldstein
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Diane S. Lidke
- Department of Pathology, University of New Mexico, Albuquerque, NM 87131
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131
| | - Bridget S. Wilson
- Department of Pathology, University of New Mexico, Albuquerque, NM 87131
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131
| | - S. Gnanakaran
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545
| |
Collapse
|
237
|
Morris R, Kershaw NJ, Babon JJ. The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Sci 2019; 27:1984-2009. [PMID: 30267440 DOI: 10.1002/pro.3519] [Citation(s) in RCA: 494] [Impact Index Per Article: 98.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 09/24/2018] [Accepted: 09/24/2018] [Indexed: 12/21/2022]
Abstract
More than 50 cytokines signal via the JAK/STAT pathway to orchestrate hematopoiesis, induce inflammation and control the immune response. Cytokines are secreted glycoproteins that act as intercellular messengers, inducing proliferation, differentiation, growth, or apoptosis of their target cells. They act by binding to specific receptors on the surface of target cells and switching on a phosphotyrosine-based intracellular signaling cascade initiated by kinases then propagated and effected by SH2 domain-containing transcription factors. As cytokine signaling is proliferative and often inflammatory, it is tightly regulated in terms of both amplitude and duration. Here we review molecular details of the cytokine-induced signaling cascade and describe the architectures of the proteins involved, including the receptors, kinases, and transcription factors that initiate and propagate signaling and the regulatory proteins that control it.
Collapse
Affiliation(s)
- Rhiannon Morris
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, 3050, Victoria, Australia
| | - Nadia J Kershaw
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, 3050, Victoria, Australia
| | - Jeffrey J Babon
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, 3050, Victoria, Australia
| |
Collapse
|
238
|
Liu X, Huang J, Liu L, Liu R. MPZL1 is highly expressed in advanced gallbladder carcinoma and promotes the aggressive behavior of human gallbladder carcinoma GBC‑SD cells. Mol Med Rep 2019; 20:2725-2733. [PMID: 31322261 PMCID: PMC6691252 DOI: 10.3892/mmr.2019.10506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 05/09/2019] [Indexed: 01/17/2023] Open
Abstract
Myelin protein 0‑like 1 (MPZL1) has been reported to have a role in hepatocellular carcinoma. However, to the best of our knowledge, there have been no studies on the function and molecular mechanism of MPZL1 gene in gallbladder carcinoma. The present study confirmed that MPZL1 was upregulated in four gallbladder carcinoma tissues according to the mRNA microarray analysis. The results of the immunohistochemical analysis of tissues from 82 patients with gallbladder carcinoma demonstrated that patients with advanced tumor stages (both T and N stage) had higher positive expression of MPZL1. Moreover, a total of 20 cases of gallbladder carcinoma and matched paired paracarcinoma tissues along with 20 samples of healthy gallbladder tissue from patients with cholecystitis were analyzed using reverse transcription‑quantitative PCR and western blotting. The results demonstrated that the expression of MPZL1 in gallbladder carcinoma tissues was significantly higher than that of paired paracarcinoma tissues and randomly matched normal gallbladder epithelial tissues. According to the Tumor‑Node‑Metastasis classification, the expression level of MPZL1 protein in stage IV gallbladder carcinoma was significantly higher than that in stage III gallbladder carcinoma. The enhanced expression of MPZL1 gene appeared to improve the migration ability of GBC‑SD cells. Conversely, GBC‑SD cells that transfected with MPZL1 siRNA exhibited decreased migration ability. The results of proliferation experiments showed that the knockdown of MPZL1 siRNA caused impairments in GBC‑SD cell proliferation. On the contrary, the overexpression of MPZL1 increased the proliferation ability of GBC‑SD cells. The results of flow cytometry analyses indicated that the upregulation of MPZL1 had an anti‑apoptotic effect on GBC‑SD cells. In conclusion, the present study showed that the expression and protein levels of MPZL1 were significantly higher in gallbladder carcinoma tissues, especially in patients diagnosed with advanced tumor stages. Overexpression of MPZL1 may have promoted the invasion, metastasis, proliferation and survival of GBC‑SD cells.
Collapse
Affiliation(s)
- Xiaolei Liu
- Department of Hepato‑Pancreato‑Biliary Surgical Oncology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Jia Huang
- Department of General Surgery, China‑Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Liguo Liu
- Department of General Surgery, China‑Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Rong Liu
- Department of Hepato‑Pancreato‑Biliary Surgical Oncology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
239
|
Agius MP, Ko KS, Johnson TK, Kwarcinski FE, Phadke S, Lachacz EJ, Soellner MB. Selective Proteolysis to Study the Global Conformation and Regulatory Mechanisms of c-Src Kinase. ACS Chem Biol 2019; 14:1556-1563. [PMID: 31287657 PMCID: PMC7254491 DOI: 10.1021/acschembio.9b00306] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Protein kinase pathways are traditionally mapped by monitoring downstream phosphorylation. Meanwhile, the noncatalytic functions of protein kinases remain under-appreciated as critical components of kinase signaling. c-Src is a protein kinase known to have noncatalytic signaling function important in healthy and disease cell signaling. Large conformational changes in the regulatory domains regulate c-Src's noncatalytic functions. Herein, we demonstrate that changes in the global conformation of c-Src can be monitored using a selective proteolysis methodology. Further, we use this methodology to investigate changes in the global conformation of several clinical and nonclinical mutations of c-Src. Significantly, we identify a novel activating mutation observed clinically, W121R, that can escape down-regulation mechanisms. Our methodology can be expanded to monitor the global conformation of other tyrosine kinases, including c-Abl, and represents an important tool toward the elucidation of the noncatalytic functions of protein kinases.
Collapse
Affiliation(s)
- Michael P. Agius
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI
| | - Kristin S. Ko
- Department of Chemistry, University of Michigan, Ann Arbor, MI
| | - Taylor K. Johnson
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI
| | | | - Sameer Phadke
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Eric J. Lachacz
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Matthew B. Soellner
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Department of Chemistry, University of Michigan, Ann Arbor, MI
| |
Collapse
|
240
|
Chang LY, Fan SMY, Liao YC, Wang WH, Chen YJ, Lin SJ. Proteomic Analysis Reveals Anti-Fibrotic Effects of Blue Light Photobiomodulation on Fibroblasts. Lasers Surg Med 2019; 52:358-372. [PMID: 31321797 DOI: 10.1002/lsm.23137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND OBJECTIVES This study was aimed at determining the effects of blue light photobiomodulation on primary adult mouse dermal fibroblasts (AMDFs) and the associated signaling pathways. STUDY DESIGN/MATERIALS AND METHODS Cultured AMDFs from adult C57BL/6 mice were irradiated by blue light from a light-emitting diode (wavelength = 463 ± 50 nm; irradiance = 5 mW/cm2 ; energy density = 4-8 J/cm2 ). The cells were analyzed using mass spectrometry for proteomics/phosphoproteomics, AlamarBlue assay for mitochondrial activity, time-lapse video for cell migration, quantitative polymerase chain reaction for gene expression, and immunofluorescence for protein expression. RESULTS Proteomic/phosphoproteomic analysis showed inhibition of extracellular signal-regulated kinases/mammalian target of rapamycin and casein kinase 2 pathways, cell motility-related networks, and multiple metabolic processes, including carbon metabolism, biosynthesis of amino acid, glycolysis/gluconeogenesis, and the pentose phosphate pathway. Functional analysis demonstrated inhibition of mitochondrial activities, cell migration, and mitosis. Expression of growth promoting insulin-like growth factor 1 and fibrosis-related genes, including transforming growth factor β1 (TGFβ1) and collagen type 1 ɑ2 chain diminished. Protein expression of α-smooth muscle actin, an important regulator of myofibroblast functions, was also suppressed. CONCLUSIONS Low-level blue light exerted suppressive effects on AMDFs, including suppression of mitochondrial activity, metabolism, cell motility, proliferation, TGFβ1 levels, and collagen I production. Low-level blue light can be a potential treatment for the prevention and reduction of tissue fibrosis, such as hypertrophic scar and keloids. Lasers Surg. Med. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lo-Yu Chang
- School of Medicine, College of Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Sabrina Mai-Yi Fan
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Yen-Chen Liao
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan.,Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd, Taipei 115, Taiwan
| | - Wei-Hung Wang
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Yu-Ju Chen
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan.,Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd, Taipei 115, Taiwan
| | - Sung-Jan Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan.,Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, No. 7, Chung-Shan South Road, Taipei 100, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, No. 7, Chung-Shan South Road, Taipei 100, Taiwan
| |
Collapse
|
241
|
Identification of Protein Phosphorylation Sites by Advanced LC-ESI-MS/MS Methods. Methods Mol Biol 2019. [PMID: 31256379 DOI: 10.1007/978-1-4939-9055-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Phosphorylation, the process by which a phosphate group is attached to a preexisting protein, is an evolutionarily and metabolically cheap way to change the protein's surface and properties. It is presumably for that reason that it is the most widespread protein modification: An estimated 10-30% of all proteins are subject to phosphorylation.MS-based methods are the methods of choice for the identification of phosphorylation sites; however biochemical pre-fractionation and enrichment protocols will be needed to produce suitable samples in the case of low-stoichiometry phosphorylation. Using emerging MS-based technology, the elucidation of the "phosphoproteome," a comprehensive inventory of phosphorylation sites, will become a realistic goal. However, validating these findings in a cellular context and defining their biological meaning remains a daunting task, which will inevitably require extensive and time-consuming additional biological research.
Collapse
|
242
|
Bellinzoni M, Wehenkel AM, Durán R, Alzari PM. Novel mechanistic insights into physiological signaling pathways mediated by mycobacterial Ser/Thr protein kinases. Microbes Infect 2019; 21:222-229. [PMID: 31254628 DOI: 10.1016/j.micinf.2019.06.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/02/2019] [Indexed: 12/11/2022]
Abstract
Protein phosphorylation is known to be one of the keystones of signal sensing and transduction in all living organisms. Once thought to be essentially confined to the eukaryotic kingdoms, reversible phosphorylation on serine, threonine and tyrosine residues, has now been shown to play a major role in many prokaryotes, where the number of Ser/Thr protein kinases (STPKs) equals or even exceeds that of two component systems. Mycobacterium tuberculosis, the etiological agent of tuberculosis, is one of the most studied organisms for the role of STPK-mediated signaling in bacteria. Driven by the interest and tractability of these enzymes as potential therapeutic targets, extensive studies revealed the remarkable conservation of protein kinases and their cognate phosphatases across evolution, and their involvement in bacterial physiology and virulence. Here, we present an overview of the current knowledge of mycobacterial STPKs structures and kinase activation mechanisms, and we then focus on PknB and PknG, two well-characterized STPKs that are essential for the intracellular survival of the bacillus. We summarize the mechanistic evidence that links PknB to the regulation of peptidoglycan synthesis in cell division and morphogenesis, and the major findings that establishes PknG as a master regulator of central carbon and nitrogen metabolism. Two decades after the discovery of STPKs in M. tuberculosis, the emerging landscape of O-phosphosignaling is starting to unveil how eukaryotic-like kinases can be engaged in unique, non-eukaryotic-like, signaling mechanisms in mycobacteria.
Collapse
Affiliation(s)
- Marco Bellinzoni
- Unit of Structural Microbiology, Department of Structural Biology and Chemistry, Institut Pasteur, CNRS UMR 3528 & Université Paris Diderot, 25 rue du Docteur Roux, 75724 Paris cedex 15, France
| | - Anne Marie Wehenkel
- Unit of Structural Microbiology, Department of Structural Biology and Chemistry, Institut Pasteur, CNRS UMR 3528 & Université Paris Diderot, 25 rue du Docteur Roux, 75724 Paris cedex 15, France
| | - Rosario Durán
- Unidad de Bioquímica y Proteómica Analíticas, Institut Pasteur de Montevideo, Instituto de Investigaciones Biológicas Clemente Estable, Mataojo 2020, Montevideo 11400, Uruguay
| | - Pedro M Alzari
- Unit of Structural Microbiology, Department of Structural Biology and Chemistry, Institut Pasteur, CNRS UMR 3528 & Université Paris Diderot, 25 rue du Docteur Roux, 75724 Paris cedex 15, France.
| |
Collapse
|
243
|
Wang Y, Yan H, Qiu Z, Hu B, Zeng B, Zhong C, Fan C. Comprehensive Analysis of SnRK Gene Family and their Responses to Salt Stress in Eucalyptus grandis. Int J Mol Sci 2019; 20:E2786. [PMID: 31174407 PMCID: PMC6600528 DOI: 10.3390/ijms20112786] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/02/2019] [Accepted: 06/04/2019] [Indexed: 11/17/2022] Open
Abstract
The sucrose non-fermentation-related protein kinase (SnRK) is a kind of Ser/Thr protein kinase, which plays a crucial role in plant stress response by phosphorylating the target protein to regulate the interconnection of various signaling pathways. However, little is known about the SnRK family in Eucalyptus grandis. Thirty-four putative SnRK sequences were identified in E. grandis and divided into three subgroups (SnRK1, SnRK2 and SnRK3) based on phylogenetic analysis and the type of domain. Chromosome localization showed that SnRK family members are unevenly distributed in the remaining 10 chromosomes, with the notable exception of chromosome 11. Gene structure analysis reveal that 10 of the 24 SnRK3 genes contained no introns. Moreover, conserved motif analyses showed that SnRK sequences belonged to the same subgroup that contained the same motif type of motif. The Ka/Ks ratio of 17 paralogues suggested that the EgrSnRK gene family underwent a purifying selection. The upstream region of EgrSnRK genes enriched with different type and numbers of cis-elements indicated that EgrSnRK genes are likely to play a role in the response to diverse stresses. Quantitative real-time PCR showed that the majority of the SnRK genes were induced by salt treatment. Genome-wide analyses and expression pattern analyses provided further understanding on the function of the SnRK family in the stress response to different environmental salt concentrations.
Collapse
Affiliation(s)
- Yujiao Wang
- Key Laboratory of State Forestry Administration on Tropical Forest Research, Research Institute of Tropical Forestry, Chinese Academy of Forestry, Guangzhou 510520, China.
- .Key Laboratory of Forest Genetics and Biotechnology, Ministry of Education of China, Co-Innovation Center for the Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China.
| | - Huifang Yan
- Key Laboratory of State Forestry Administration on Tropical Forest Research, Research Institute of Tropical Forestry, Chinese Academy of Forestry, Guangzhou 510520, China.
| | - Zhenfei Qiu
- Key Laboratory of State Forestry Administration on Tropical Forest Research, Research Institute of Tropical Forestry, Chinese Academy of Forestry, Guangzhou 510520, China.
| | - Bing Hu
- Key Laboratory of State Forestry Administration on Tropical Forest Research, Research Institute of Tropical Forestry, Chinese Academy of Forestry, Guangzhou 510520, China.
| | - Bingshan Zeng
- Key Laboratory of State Forestry Administration on Tropical Forest Research, Research Institute of Tropical Forestry, Chinese Academy of Forestry, Guangzhou 510520, China.
| | - Chonglu Zhong
- Key Laboratory of State Forestry Administration on Tropical Forest Research, Research Institute of Tropical Forestry, Chinese Academy of Forestry, Guangzhou 510520, China.
| | - Chunjie Fan
- Key Laboratory of State Forestry Administration on Tropical Forest Research, Research Institute of Tropical Forestry, Chinese Academy of Forestry, Guangzhou 510520, China.
| |
Collapse
|
244
|
Wozniak JM, Gonzalez DJ. PTMphinder: an R package for PTM site localization and motif extraction from proteomic datasets. PeerJ 2019; 7:e7046. [PMID: 31198645 PMCID: PMC6555389 DOI: 10.7717/peerj.7046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/24/2019] [Indexed: 11/30/2022] Open
Abstract
Background Mass-spectrometry-based proteomics is a prominent field of study that allows for the unbiased quantification of thousands of proteins from a particular sample. A key advantage of these techniques is the ability to detect protein post-translational modifications (PTMs) and localize them to specific amino acid residues. These approaches have led to many significant findings in a wide range of biological disciplines, from developmental biology to cancer and infectious diseases. However, there is a current lack of tools available to connect raw PTM site information to biologically meaningful results in a high-throughput manner. Furthermore, many of the available tools require significant programming knowledge to implement. Results The R package PTMphinder was designed to enable researchers, particularly those with minimal programming background, to thoroughly analyze PTMs in proteomic data sets. The package contains three functions: parseDB, phindPTMs and extractBackground. Together, these functions allow users to reformat proteome databases for easier analysis, localize PTMs within full proteins, extract motifs surrounding the identified sites and create proteome-specific motif backgrounds for statistical purposes. Beta-testing of this R package has demonstrated its simplicity and ease of integration with existing tools. Conclusion PTMphinder empowers researchers to fully analyze and interpret PTMs derived from proteomic data. This package is simple enough for researchers with limited programming experience to understand and implement. The data produced from this package can inform subsequent research by itself and also be used in conjunction with other tools, such as motif-x, for further analysis.
Collapse
Affiliation(s)
- Jacob M Wozniak
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States of America.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States of America
| | - David J Gonzalez
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States of America.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States of America
| |
Collapse
|
245
|
Li S, Chen X, Wang N, Li J, Feng Y, Sun J. Identification and characterization of ecto-nucleoside triphosphate diphosphohydrolase 1 (CD39) involved in regulating extracellular ATP-mediated innate immune responses in Japanese flounder (Paralichthys olivaceus). Mol Immunol 2019; 112:10-21. [PMID: 31075558 DOI: 10.1016/j.molimm.2019.04.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/28/2019] [Accepted: 04/28/2019] [Indexed: 12/17/2022]
Abstract
Extracellular adenosine triphosphate (eATP), released following inflammatory stimulation or infection, is a potent signaling molecule in activating innate immune responses in fish. However, the regulation of eATP-mediated innate immunity in fish remains unknown. Ecto-nucleoside triphosphate diphosphohydrolase 1 (CD39) is a critical molecular switch for controlling the ATP levels in the extracellular space. CD39 plays a key role in regulating eATP-activated innate immune responses through the phosphohydrolysis of pro-inflammatory eATP to inactive AMP. Here, we identified and characterized a CD39 homolog (namely, poCD39) in the Japanese flounder Paralichthys olivaceus and analyzed its regulatory role in eATP-mediated innate immunity. Real-time quantitative PCR analysis revealed that poCD39 is ubiquitously present in all tested normal tissues with dominant expression in enriched Japanese flounder head kidney macrophages (HKMs). Immune challenge experiments demonstrated that poCD39 expression was upregulated by inflammatory stimulation and Edwardsiella tarda infection. Biochemical and immunofluorescence analysis revealed that poCD39 is a functional glycosylated membrane protein for the hydrolysis of eATP. Inhibition of poCD939 activity with the ecto-NTPDase inhibitor ARL 67156 resulted in increased IL-1beta gene expression and ROS production in Japanese flounder HKMs. In contrast, overexpression of poCD39 in Japanese flounder FG-9307 cells reduced eATP-induced pro-inflammatory cytokine IL-1beta gene expression. Finally, poCD39 expression was significantly induced by eATP stimulation in the HKMs, suggesting that eATP may provide a feedback mechanism for transcriptional regulation of fish CD39. Taken together, we identified and characterized a functional fish CD39 protein involved in regulating eATP-mediated innate immune responses in fish.
Collapse
Affiliation(s)
- Shuo Li
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China.
| | - Xiaoli Chen
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China
| | - Nan Wang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China
| | - Jiafang Li
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China
| | - Yu Feng
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China
| | - Jinsheng Sun
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China.
| |
Collapse
|
246
|
Gomes ID, Pflum MKH. Optimal Substrate-Trapping Mutants to Discover Substrates of HDAC1. Chembiochem 2019; 20:1444-1449. [PMID: 30701667 DOI: 10.1002/cbic.201800797] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Indexed: 11/11/2022]
Abstract
Histone deacetylase 1 (HDAC1) regulates transcription by deacetylating histones. In addition to histones, several non-histone proteins are HDAC1 substrates, which suggests a role for HDAC1 beyond epigenetics. Unfortunately, the identification of non-histone substrates has been largely serendipitous, which makes full characterization of HDAC1 functions difficult. To overcome this challenge, inactive "trapping" mutants were recently developed to identify HDAC1 substrates. To optimize substrate trapping, the relative trapping abilities of 17 inactive HDAC1 mutants was assessed. HDAC1 H141A, F150A, and C151A showed strong binding to substrates LSD1 and p53. Interestingly, each mutant preferentially trapped a different substrate. By combining several inactive mutants, the trapping strategy will facilitate the discovery of new HDAC1 substrates and shed light on the variety of HDAC1-related functions in cell biology.
Collapse
Affiliation(s)
- Inosha D Gomes
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI, 48202, USA
| | - Mary Kay H Pflum
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI, 48202, USA
| |
Collapse
|
247
|
Bellinzoni M, Wehenkel AM, Durán R, Alzari PM. Novel mechanistic insights into physiological signaling pathways mediated by mycobacterial Ser/Thr protein kinases. Genes Immun 2019; 20:383-393. [DOI: 10.1038/s41435-019-0069-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/02/2019] [Indexed: 12/16/2022]
|
248
|
Gao Y, Lee H, Kwon OK, Cheng Z, Tan M, Kim K, Lee S. Profiling of Histidine Phosphoproteome in
Danio rerio
by TiO
2
Enrichment. Proteomics 2019; 19:e1800471. [DOI: 10.1002/pmic.201800471] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/25/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Yan Gao
- BK21 Plus KNU Multi‐Omics based Creative Drug Research TeamCollege of Pharmacy and Research Institute of Pharmaceutical SciencesKyungpook National University Daegu 41566 Republic of Korea
| | - Hyojin Lee
- Department of Energy and Environmental Engineering and Department of Environmental EngineeringSeoul National University of Science and Technology Seoul 01811 Republic of Korea
| | - Oh Kwang Kwon
- BK21 Plus KNU Multi‐Omics based Creative Drug Research TeamCollege of Pharmacy and Research Institute of Pharmaceutical SciencesKyungpook National University Daegu 41566 Republic of Korea
| | - Zhongyi Cheng
- Jingjie PTM BioLab (Hangzhou) Co. Ltd. Hangzhou 310018 China
| | - Minjia Tan
- Shanghai Institute of Materia MedicaChinese Academy of Sciences Shanghai 201203 China
| | - Ki‐Tae Kim
- Department of Energy and Environmental Engineering and Department of Environmental EngineeringSeoul National University of Science and Technology Seoul 01811 Republic of Korea
- Jingjie PTM BioLab (Hangzhou) Co. Ltd. Hangzhou 310018 China
| | - Sangkyu Lee
- BK21 Plus KNU Multi‐Omics based Creative Drug Research TeamCollege of Pharmacy and Research Institute of Pharmaceutical SciencesKyungpook National University Daegu 41566 Republic of Korea
| |
Collapse
|
249
|
Hussain D, Najam-ul-Haq M, Majeed S, Musharraf SG, Lu Q, He X, Feng YQ. Facile liquid-phase deposition synthesis of titania-coated magnetic sporopollenin for the selective capture of phosphopeptides. Anal Bioanal Chem 2019; 411:3373-3382. [DOI: 10.1007/s00216-019-01811-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/24/2019] [Accepted: 03/26/2019] [Indexed: 12/20/2022]
|
250
|
Suskiewicz MJ, Hajdusits B, Beveridge R, Heuck A, Vu LD, Kurzbauer R, Hauer K, Thoeny V, Rumpel K, Mechtler K, Meinhart A, Clausen T. Structure of McsB, a protein kinase for regulated arginine phosphorylation. Nat Chem Biol 2019; 15:510-518. [PMID: 30962626 DOI: 10.1038/s41589-019-0265-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 03/05/2019] [Indexed: 11/09/2022]
Abstract
Protein phosphorylation regulates key processes in all organisms. In Gram-positive bacteria, protein arginine phosphorylation plays a central role in protein quality control by regulating transcription factors and marking aberrant proteins for degradation. Here, we report structural, biochemical, and in vivo data of the responsible kinase, McsB, the founding member of an arginine-specific class of protein kinases. McsB differs in structure and mechanism from protein kinases that act on serine, threonine, and tyrosine residues and instead has a catalytic domain related to that of phosphagen kinases (PhKs), metabolic enzymes that phosphorylate small guanidino compounds. In McsB, the PhK-like phosphotransferase domain is structurally adapted to target protein substrates and is accompanied by a novel phosphoarginine (pArg)-binding domain that allosterically controls protein kinase activity. The identification of distinct pArg reader domains in this study points to a remarkably complex signaling system, thus challenging simplistic views of bacterial protein phosphorylation.
Collapse
Affiliation(s)
- Marcin J Suskiewicz
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Bence Hajdusits
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Rebecca Beveridge
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Alexander Heuck
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Lam Dai Vu
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria.,VIB/UGent, Ghent, Belgium
| | - Robert Kurzbauer
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Katja Hauer
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | | - Klaus Rumpel
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Karl Mechtler
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria.,Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Anton Meinhart
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Tim Clausen
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria. .,Medical University of Vienna, Vienna BioCenter (VBC), Vienna, Austria.
| |
Collapse
|