301
|
Mann A, Moulédous L, Froment C, O'Neill PR, Dasgupta P, Günther T, Brunori G, Kieffer BL, Toll L, Bruchas MR, Zaveri NT, Schulz S. Agonist-selective NOP receptor phosphorylation correlates in vitro and in vivo and reveals differential post-activation signaling by chemically diverse agonists. Sci Signal 2019; 12:12/574/eaau8072. [PMID: 30914485 DOI: 10.1126/scisignal.aau8072] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Agonists of the nociceptin/orphanin FQ opioid peptide (NOP) receptor, a member of the opioid receptor family, are under active investigation as novel analgesics, but their modes of signaling are less well characterized than those of other members of the opioid receptor family. Therefore, we investigated whether different NOP receptor ligands showed differential signaling or functional selectivity at the NOP receptor. Using newly developed phosphosite-specific antibodies to the NOP receptor, we found that agonist-induced NOP receptor phosphorylation occurred primarily at four carboxyl-terminal serine (Ser) and threonine (Thr) residues, namely, Ser346, Ser351, Thr362, and Ser363, and proceeded with a temporal hierarchy, with Ser346 as the first site of phosphorylation. G protein-coupled receptor kinases 2 and 3 (GRK2/3) cooperated during agonist-induced phosphorylation, which, in turn, facilitated NOP receptor desensitization and internalization. A comparison of structurally distinct NOP receptor agonists revealed dissociation in functional efficacies between G protein-dependent signaling and receptor phosphorylation. Furthermore, in NOP-eGFP and NOP-eYFP mice, NOP receptor agonists induced multisite phosphorylation and internalization in a dose-dependent and agonist-selective manner that could be blocked by specific antagonists. Our study provides new tools to study ligand-activated NOP receptor signaling in vitro and in vivo. Differential agonist-selective NOP receptor phosphorylation by chemically diverse NOP receptor agonists suggests that differential signaling by NOP receptor agonists may play a role in NOP receptor ligand pharmacology.
Collapse
Affiliation(s)
- Anika Mann
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, Jena 07747, Germany.
| | - Lionel Moulédous
- Research Center on Animal Cognition, Center for Integrative Biology, Toulouse University, CNRS, UPS, 31062 Toulouse Cedex 09, France
| | - Carine Froment
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, 31077 Toulouse Cedex 04, France
| | - Patrick R O'Neill
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Pooja Dasgupta
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, Jena 07747, Germany
| | - Thomas Günther
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, Jena 07747, Germany
| | - Gloria Brunori
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Brigitte L Kieffer
- Douglas Research Center, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC H3A 1A1, Canada
| | - Lawrence Toll
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Michael R Bruchas
- Center for the Neurobiology of Addiction, Pain, and Emotion, Departments of Anesthesiology and Pharmacology, University of Washington, Seattle, WA 98195, USA
| | | | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, Jena 07747, Germany.
| |
Collapse
|
302
|
Quintana-Gallardo L, Martín-Benito J, Marcilla M, Espadas G, Sabidó E, Valpuesta JM. The cochaperone CHIP marks Hsp70- and Hsp90-bound substrates for degradation through a very flexible mechanism. Sci Rep 2019; 9:5102. [PMID: 30911017 PMCID: PMC6433865 DOI: 10.1038/s41598-019-41060-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 01/22/2019] [Indexed: 11/26/2022] Open
Abstract
Some molecular chaperones are involved not only in assisting the folding of proteins but also, given appropriate conditions, in their degradation. This is the case for Hsp70 and Hsp90 which, in concert with the cochaperone CHIP, direct their bound substrate to degradation through ubiquitination. We generated complexes between the chaperones (Hsp70 or Hsp90), the cochaperone CHIP and, as substrate, a p53 variant containing the GST protein (p53-TMGST). Both ternary complexes (Hsp70:p53-TMGST:CHIP and Hsp90:p53-TMGST:CHIP) ubiquitinated the substrate at a higher efficiency than in the absence of the chaperones. The 3D structures of the two complexes, obtained using a combination of cryoelectron microscopy and crosslinking mass spectrometry, showed the substrate located between the chaperone and the cochaperone, suggesting a ubiquitination mechanism in which the chaperone-bound substrate is presented to CHIP. These complexes are inherently flexible, which is important for the ubiquitination process.
Collapse
Affiliation(s)
| | | | - Miguel Marcilla
- Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049, Madrid, Spain
| | - Guadalupe Espadas
- Proteomics Unit, Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Proteomics Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Eduard Sabidó
- Proteomics Unit, Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Proteomics Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | | |
Collapse
|
303
|
Wang Y, Zhang SP, Zhang MY, Kempher ML, Guo DD, Han JT, Tao X, Wu Y, Zhang LQ, He YX. The antitoxin MqsA homologue in Pseudomonas fluorescens 2P24 has a rewired regulatory circuit through evolution. Environ Microbiol 2019; 21:1740-1756. [PMID: 30680880 DOI: 10.1111/1462-2920.14538] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 01/22/2019] [Indexed: 12/01/2022]
Abstract
The mqsRA operon encodes a toxin-antitoxin pair that was characterized to participate in biofilm and persister cell formation in Escherichia coli. Notably, the antitoxin MqsA possesses a C-terminal DNA-binding domain that recognizes the [5'-AACCT(N)2-4 AGGTT-3'] motif and acts as a transcriptional regulator controlling multiple genes including the general stress response regulator RpoS. However, it is unknown how the transcriptional circuits of MqsA homologues have changed in bacteria over evolutionary time. Here, we found mqsA in Pseudomonas fluorescens (PfmqsA) is acquired through horizontal gene transfer and binds to a slightly different motif [5'-TACCCT(N)3 AGGGTA-3'], which exists upstream of the PfmqsRA operon. Interestingly, an adjacent GntR-type transcriptional regulator, which was termed AgtR, is under negative control of PfMqsA. It was further demonstrated that PfMqsA reduces production of biofilm components through AgtR, which directly regulates the pga and fap operons involved in the synthesis of extracellular polymeric substances. Moreover, through quantitative proteomics analysis, we showed AgtR is a highly pleiotropic regulator that influences up to 252 genes related to diverse processes including chemotaxis, oxidative phosphorylation and carbon and nitrogen metabolism. Taken together, our findings suggest the rewired regulatory circuit of PfMqsA influences diverse physiological aspects of P. fluorescens 2P24 via the newly characterized AgtR.
Collapse
Affiliation(s)
- Yong Wang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Si-Ping Zhang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.,School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Meng-Yuan Zhang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.,School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Megan L Kempher
- Institute for Environmental Genomics, Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Ding-Ding Guo
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Jian-Ting Han
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Xuanyu Tao
- Institute for Environmental Genomics, Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Yi Wu
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Li-Qun Zhang
- Department of Plant Pathology, China Agricultural University, Beijing, China
| | - Yong-Xing He
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
304
|
Shen ZQ, Shi B, Wang TR, Jiao J, Shang XJ, Wu QJ, Zhou YM, Cao TF, Du Q, Wang XX, Li D. Characterization of the Sperm Proteome and Reproductive Outcomes with in Vitro, Fertilization after a Reduction in Male Ejaculatory Abstinence Period. Mol Cell Proteomics 2019; 18:S109-S117. [PMID: 30126978 PMCID: PMC6427236 DOI: 10.1074/mcp.ra117.000541] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 08/03/2018] [Indexed: 12/22/2022] Open
Abstract
Semen samples from men after a short ejaculatory abstinence show improved sperm quality and result in increased pregnancy rates, but the underlying mechanisms remain unclear. Herein, we report that ejaculates from short (1-3 h) compared with long (3-7 days) periods of abstinence showed increases in motile sperm count, sperm vitality, normal sperm morphology, acrosome reaction capacity, total antioxidant capacity, sperm mitochondrial membrane potential, high DNA stainability, and a decrease in the sperm DNA fragmentation index (p, < 0.05). Sperm proteomic analysis showed 322 differentially expressed proteins (minimal fold change of ±1.5 or greater and p, < 0.05), with 224 upregulated and 98 downregulated. These differentially expressed proteins are profoundly involved in specific cellular processes, such as motility and capacitation, oxidative stress, and metabolism. Interestingly, protein trimethyllysine modification was increased, and butyryllysine, propionyllysine, and malonyllysine modifications were decreased in ejaculates from a short versus, long abstinence (p, < 0.05). Finally, the rates of implantation, clinical pregnancy, and live births from in vitro, fertilization treatments were significantly increased in semen samples after a short abstinence. Our study provides preliminary mechanistic insights into improved sperm quality and pregnancy outcomes associated with spermatozoa retrieved after a short ejaculatory abstinence.
Collapse
Affiliation(s)
- Zi-Qi Shen
- From the ‡Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Bei Shi
- Department of Physiology, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Tian-Ren Wang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06520, USA
| | - Jiao Jiao
- From the ‡Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xue-Jun Shang
- Department of Andrology, Jinling Hospital Affiliated to Nanjing University School of Medicine, Nanjing 210002, China
| | - Qi-Jun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yi-Ming Zhou
- ‡Department of Medicine, Brigham and Women's Hospital, Harvard Institutes of Medicine, Harvard Medical School, Boston, MA 02115
| | - Tie-Feng Cao
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06520, USA
| | - Qiang Du
- From the ‡Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiu-Xia Wang
- From the ‡Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China;.
| | - Da Li
- From the ‡Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China;.
| |
Collapse
|
305
|
Arsenic Trioxide and (-)-Gossypol Synergistically Target Glioma Stem-Like Cells via Inhibition of Hedgehog and Notch Signaling. Cancers (Basel) 2019; 11:cancers11030350. [PMID: 30871073 PMCID: PMC6468469 DOI: 10.3390/cancers11030350] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/28/2019] [Accepted: 03/07/2019] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma is one of the deadliest malignancies and is virtually incurable. Accumulating evidence indicates that a small population of cells with a stem-like phenotype is the major culprit of tumor recurrence. Enhanced DNA repair capacity and expression of stemness marker genes are the main characteristics of these cells. Elimination of this population might delay or prevent tumor recurrence following radiochemotherapy. The aim of this study was to analyze whether interference with the Hedgehog signaling (Hh) pathway or combined Hh/Notch blockade using small-molecule inhibitors can efficiently target these cancer stem cells and sensitize them to therapy. Using tumor sphere lines and primary patient-derived glioma cultures we demonstrate that the Hh pathway inhibitor GANT61 (GANT) and the arsenic trioxide (ATO)-mediated Hh/Notch inhibition are capable to synergistically induce cell death in combination with the natural anticancer agent (−)-Gossypol (Gos). Only ATO in combination with Gos also strongly decreased stemness marker expression and prevented sphere formation and recovery. These synergistic effects were associated with distinct proteomic changes indicating diminished DNA repair and markedly reduced stemness. Finally, using an organotypic brain slice transplantation model, we show that combined ATO/Gos treatment elicits strong growth inhibition or even complete elimination of tumors. Collectively, our data show for the first time that ATO and Gos, two drugs that can be used in the clinic, represent a promising targeted therapy approach for the synergistic elimination of glioma stem-like cells.
Collapse
|
306
|
Naamati A, Williamson JC, Greenwood EJ, Marelli S, Lehner PJ, Matheson NJ. Functional proteomic atlas of HIV infection in primary human CD4+ T cells. eLife 2019; 8:41431. [PMID: 30857592 PMCID: PMC6414203 DOI: 10.7554/elife.41431] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/10/2019] [Indexed: 12/19/2022] Open
Abstract
Viruses manipulate host cells to enhance their replication, and the identification of cellular factors targeted by viruses has led to key insights into both viral pathogenesis and cell biology. In this study, we develop an HIV reporter virus (HIV-AFMACS) displaying a streptavidin-binding affinity tag at the surface of infected cells, allowing facile one-step selection with streptavidin-conjugated magnetic beads. We use this system to obtain pure populations of HIV-infected primary human CD4+ T cells for detailed proteomic analysis, and quantitate approximately 9000 proteins across multiple donors on a dynamic background of T cell activation. Amongst 650 HIV-dependent changes (q < 0.05), we describe novel Vif-dependent targets FMR1 and DPH7, and 192 proteins not identified and/or regulated in T cell lines, such as ARID5A and PTPN22. We therefore provide a high-coverage functional proteomic atlas of HIV infection, and a mechanistic account of host factors subverted by the virus in its natural target cell.
Collapse
Affiliation(s)
- Adi Naamati
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - James C Williamson
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Edward Jd Greenwood
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Sara Marelli
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Paul J Lehner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
307
|
Curto P, Santa C, Allen P, Manadas B, Simões I, Martinez JJ. A Pathogen and a Non-pathogen Spotted Fever Group Rickettsia Trigger Differential Proteome Signatures in Macrophages. Front Cell Infect Microbiol 2019; 9:43. [PMID: 30895174 PMCID: PMC6414445 DOI: 10.3389/fcimb.2019.00043] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/11/2019] [Indexed: 12/13/2022] Open
Abstract
We have previously reported that Rickettsia conorii and Rickettsia montanensis have distinct intracellular fates within THP-1 macrophages, suggesting that the ability to proliferate within macrophages may be a distinguishable factor between pathogenic and non-pathogenic Spotted fever group (SFG) members. To start unraveling the molecular mechanisms underlying the capacity (or not) of SFG Rickettsia to establish their replicative niche in macrophages, we have herein used quantitative proteomics by SWATH-MS to profile the alterations resulted by the challenge of THP-1 macrophages with R. conorii and R. montanensis. We show that the pathogenic, R. conorii, and the non-pathogenic, R. montanensis, member of SFG Rickettsia trigger differential proteomic signatures in macrophage-like cells upon infection. R. conorii specifically induced the accumulation of several enzymes of the tricarboxylic acid cycle, oxidative phosphorylation, fatty acid β-oxidation, and glutaminolysis, as well as of several inner and outer membrane mitochondrial transporters. These results suggest a profound metabolic rewriting of macrophages by R. conorii toward a metabolic signature of an M2-like, anti-inflammatory activation program. Moreover, several subunits forming the proteasome and immunoproteasome are found in lower abundance upon infection with both rickettsial species, which may help bacteria to escape immune surveillance. R. conorii-infection specifically induced the accumulation of several host proteins implicated in protein processing and quality control in ER, suggesting that this pathogenic Rickettsia may be able to increase the ER protein folding capacity. This work reveals novel aspects of macrophage-Rickettsia interactions, expanding our knowledge of how pathogenic rickettsiae explore host cells to their advantage.
Collapse
Affiliation(s)
- Pedro Curto
- PhD Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Cátia Santa
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Paige Allen
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Bruno Manadas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Isaura Simões
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Juan J. Martinez
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
308
|
Charkoftaki G, Thompson DC, Golla JP, Garcia-Milian R, Lam TT, Engel J, Vasiliou V. Integrated multi-omics approach reveals a role of ALDH1A1 in lipid metabolism in human colon cancer cells. Chem Biol Interact 2019; 304:88-96. [PMID: 30851239 DOI: 10.1016/j.cbi.2019.02.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/16/2019] [Accepted: 02/28/2019] [Indexed: 01/06/2023]
Affiliation(s)
- Georgia Charkoftaki
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, 06520, USA
| | - David C Thompson
- Department of Clinical Pharmacy, Skaggs School of Pharmacy & Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Jaya Prakash Golla
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, 06520, USA
| | - Rolando Garcia-Milian
- Bioinformatics Support Program, Cushing/Whitney Medical Library, Yale School of Medicine, New Haven, CT, 06250, USA
| | - TuKiet T Lam
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, 06510, USA; Yale MS & Proteomics Resource, WM Keck Biotechnology Resource Laboratory, New Haven, CT, 06510, USA
| | - Jasper Engel
- Biometris, Wageningen University & Research, Wagenigen, the Netherlands
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
309
|
Li Y, Jia Y, Bian Y, Tong H, Qu J, Wang K, Wan XP. Autocrine motility factor promotes endometrial cancer progression by targeting GPER-1. Cell Commun Signal 2019; 17:22. [PMID: 30836961 PMCID: PMC6402158 DOI: 10.1186/s12964-019-0336-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 02/26/2019] [Indexed: 01/01/2023] Open
Abstract
Background Autocrine motility factor (AMF) is a critical factor regulating aggressiveness of endometrial cancer (EC). Multiple pieces of evidence indicate that it is through G protein coupled estrogen receptor (GPER) signaling pathway that some growth factors promoted the migration and proliferation of tumor cells. The aim of this study is to explore the role of GPER-1 in AMF mediated regulatory mechanisms of EC recurrence and progression. Methods Real-Time Cell Analysis (RTCA) assays were performed to assess whether AMF depends on Autocrine motility factor recepter (AMFR) signaling in EC cells. A genome-wide expression microarray and Yeast Two-Hybrid assay were used to detect AMF and GPER-1 interaction in the context of AMFR depletion, and co-immunoprecipitation and immunofluorescence experiments were performed to confirm the physical interaction. Isobaric Tags for Relative and Absolute Quantification (iTRAQ) analysis was used for the identification of the target pathway activated by AMF-GPER-1 interaction. Cohorts of mice harboring xenografts derived from modified SPEC2 cell lines were treated with or without exogenous AMF to validate the results of previous experiments. Immunohistochemistry was performed to assess AMF and GPER-1 expression in endometrial cancer specimens and normal endometrium. Results Our data showed that GPER-1 binds to AMF and the formed complex translocates from the plasma membrane to the cytoplasm. Mechanistic investigations demonstrated that interaction between AMF and GPER-1 triggers phosphoinositide-3-kinase signaling and promotes EC cell growth. More importantly, through animal experiments and human tissue experiments, we found that AMF contributes to GPER-1-mediated EC progression, which is consistent with the above observations. Conclusions Our work not only delineated the regulatory mechanisms of endometrial cancer progression by AMF-GPER-1-AKT signaling cascade but also laid the foundation of targeting this pathway for treating endometrial cancer. Electronic supplementary material The online version of this article (10.1186/s12964-019-0336-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yiran Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuanhui Jia
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yiding Bian
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Huan Tong
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Junjie Qu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kai Wang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China.
| | - Xiao-Ping Wan
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
310
|
McKetney J, Runde R, Hebert AS, Salamat S, Roy S, Coon JJ. Proteomic Atlas of the Human Brain in Alzheimer's Disease. J Proteome Res 2019; 18:1380-1391. [PMID: 30735395 PMCID: PMC6480317 DOI: 10.1021/acs.jproteome.9b00004] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The brain represents one of the most divergent and critical organs in the human body. Yet, it can be afflicted by a variety of neurodegenerative diseases specifically linked to aging, about which we lack a full biomolecular understanding of onset and progression, such as Alzheimer's disease (AD). Here we provide a proteomic resource comprising nine anatomically distinct sections from three aged individuals, across a spectrum of disease progression, categorized by quantity of neurofibrillary tangles. Using state-of-the-art mass spectrometry, we identify a core brain proteome that exhibits only small variance in expression, accompanied by a group of proteins that are highly differentially expressed in individual sections and broader regions. AD affected tissue exhibited slightly elevated levels of tau protein with similar relative expression to factors associated with the AD pathology. Substantial differences were identified between previous proteomic studies of mature adult brains and our aged cohort. Our findings suggest considerable value in examining specifically the brain proteome of aged human populations from a multiregional perspective. This resource can serve as a guide, as well as a point of reference for how specific regions of the brain are affected by aging and neurodegeneration.
Collapse
Affiliation(s)
- Justin McKetney
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Rosie Runde
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison
- Department of Neuroscience, University of Wisconsin–Madison, 1111 Highland Avenue, Madison, Wisconsin 53705
| | - Alexander S Hebert
- Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI 53706
| | - Shahriar Salamat
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison
- Department of Neuroscience, University of Wisconsin–Madison, 1111 Highland Avenue, Madison, Wisconsin 53705
| | - Subhojit Roy
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison
- Department of Neuroscience, University of Wisconsin–Madison, 1111 Highland Avenue, Madison, Wisconsin 53705
| | - Joshua J. Coon
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706
- Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI 53706
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706
- Morgridge Institute for Research, Madison, WI 53706
| |
Collapse
|
311
|
Hoque A, Williamson NA, Ameen SS, Ciccotosto GD, Hossain MI, Oakhill JS, Ng DCH, Ang CS, Cheng HC. Quantitative proteomic analyses of dynamic signalling events in cortical neurons undergoing excitotoxic cell death. Cell Death Dis 2019; 10:213. [PMID: 30824683 PMCID: PMC6397184 DOI: 10.1038/s41419-019-1445-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/20/2019] [Accepted: 02/01/2019] [Indexed: 12/23/2022]
Abstract
Excitotoxicity, caused by overstimulation or dysregulation of ionotropic glutamate receptors (iGluRs), is a pathological process directing neuronal death in many neurological disorders. The aberrantly stimulated iGluRs direct massive influx of calcium ions into the affected neurons, leading to changes in expression and phosphorylation of specific proteins to modulate their functions and direct their participation in the signalling pathways that induce excitotoxic neuronal death. To define these pathways, we used quantitative proteomic approaches to identify these neuronal proteins (referred to as the changed proteins) and determine how their expression and/or phosphorylation dynamically changed in association with excitotoxic cell death. Our data, available in ProteomeXchange with identifier PXD008353, identified over 100 changed proteins exhibiting significant alterations in abundance and/or phosphorylation levels at different time points (5–240 min) in neurons after glutamate overstimulation. Bioinformatic analyses predicted that many of them are components of signalling networks directing defective neuronal morphology and functions. Among them, the well-known neuronal survival regulators including mitogen-activated protein kinases Erk1/2, glycogen synthase kinase 3 (GSK3) and microtubule-associated protein (Tau), were selected for validation by biochemical approaches, which confirmed the findings of the proteomic analysis. Bioinformatic analysis predicted Protein Kinase B (Akt), c-Jun kinase (JNK), cyclin-dependent protein kinase 5 (Cdk5), MAP kinase kinase (MEK), Casein kinase 2 (CK2), Rho-activated protein kinase (Rock) and Serum/glucocorticoid-regulated kinase 1 (SGK1) as the potential upstream kinases phosphorylating some of the changed proteins. Further biochemical investigation confirmed the predictions of sustained changes of the activation states of neuronal Akt and CK2 in excitotoxicity. Thus, future investigation to define the signalling pathways directing the dynamic alterations in abundance and phosphorylation of the identified changed neuronal proteins will help elucidate the molecular mechanism of neuronal death in excitotoxicity.
Collapse
Affiliation(s)
- Ashfaqul Hoque
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia.,Cell Signalling Research Laboratories, University of Melbourne, Parkville, VIC, 3010, Australia.,Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.,Metabolic Signalling Laboratory, St. Vincent's Institute for Medical Research, University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Nicholas A Williamson
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - S Sadia Ameen
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia.,Cell Signalling Research Laboratories, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Giuseppe D Ciccotosto
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC, 3010, Australia
| | - M Iqbal Hossain
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia.,Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St. Vincent's Institute for Medical Research, University of Melbourne, Fitzroy, VIC, 3065, Australia.,Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, 3000, Australia
| | - Dominic C H Ng
- School of Biomedical Sciences, University of Queensland, St. Lucia, QLD, Australia
| | - Ching-Seng Ang
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Heung-Chin Cheng
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia. .,Cell Signalling Research Laboratories, University of Melbourne, Parkville, VIC, 3010, Australia. .,Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
312
|
Differential protein expression of blood platelet components associated with adverse transfusion reactions. J Proteomics 2019; 194:25-36. [DOI: 10.1016/j.jprot.2018.12.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/13/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023]
|
313
|
Aranda S, Alcaine-Colet A, Blanco E, Borràs E, Caillot C, Sabidó E, Di Croce L. Chromatin capture links the metabolic enzyme AHCY to stem cell proliferation. SCIENCE ADVANCES 2019; 5:eaav2448. [PMID: 30854431 PMCID: PMC6402848 DOI: 10.1126/sciadv.aav2448] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 01/28/2019] [Indexed: 05/19/2023]
Abstract
Profiling the chromatin-bound proteome (chromatome) in a simple, direct, and reliable manner might be key to uncovering the role of yet uncharacterized chromatin factors in physiology and disease. Here, we have designed an experimental strategy to survey the chromatome of proliferating cells by using the DNA-mediated chromatin pull-down (Dm-ChP) technology. Our approach provides a global view of cellular chromatome under normal physiological conditions and enables the identification of chromatin-bound proteins de novo. Integrating Dm-ChP with genomic and functional data, we have discovered an unexpected chromatin function for adenosylhomocysteinase, a major one-carbon pathway metabolic enzyme, in gene activation. Our study reveals a new regulatory axis between the metabolic state of pluripotent cells, ribosomal protein production, and cell division during the early phase of embryo development, in which the metabolic flux of methylation reactions is favored in a local milieu.
Collapse
Affiliation(s)
- Sergi Aranda
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Anna Alcaine-Colet
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Enrique Blanco
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Eva Borràs
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Claire Caillot
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Eduard Sabidó
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Luciano Di Croce
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluis Companys 23, Barcelona 08010, Spain
| |
Collapse
|
314
|
Tsolis KC, Hamed MB, Simoens K, Koepff J, Busche T, Rückert C, Oldiges M, Kalinowski J, Anné J, Kormanec J, Bernaerts K, Karamanou S, Economou A. Secretome Dynamics in a Gram-Positive Bacterial Model. Mol Cell Proteomics 2019; 18:423-436. [PMID: 30498012 PMCID: PMC6398212 DOI: 10.1074/mcp.ra118.000899] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 11/21/2018] [Indexed: 12/11/2022] Open
Abstract
Protein secretion is a central biological process in all organisms. Most studies dissecting bacterial secretion mechanisms have focused on Gram-negative cell envelopes such as that of Escherichia coli However, proteomics analyses in Gram negatives is hampered by their outer membrane. Here we studied protein secretion in the Gram-positive bacterium Streptomyces lividans TK24, in which most of the secretome is released in the growth medium. We monitored changes of the secretome as a function of growth phase and medium. We determined distinct protein classes of "house-keeping" secreted proteins that do not change their appearance or abundance in the various media and growth phases. These comprise mainly enzymes involved in cell wall maintenance and basic transport. In addition, we detected significant abundance and content changes to a sub-set of the proteome, as a function of growth in the different media. These did not depend on the media being minimal or rich. Transcriptional regulation but not changes in export machinery components can explain some of these changes. However, additional downstream mechanisms must be important for selective secretome funneling. These observations lay the foundations of using S. lividans as a model organism to study how metabolism is linked to optimal secretion and help develop rational optimization of heterologous protein production.
Collapse
Affiliation(s)
- Konstantinos C Tsolis
- From the ‡KU Leuven, Rega Institute, Dpt of Microbiology and Immunology, Herestraat 49, B-3000 Leuven, Belgium
| | - Mohamed Belal Hamed
- From the ‡KU Leuven, Rega Institute, Dpt of Microbiology and Immunology, Herestraat 49, B-3000 Leuven, Belgium
- ‡‡Molecular Biology Dpt, National Research Centre, Dokki, Giza, Egypt
| | - Kenneth Simoens
- §KU Leuven, Bio- & chemical systems Technology, Reactor Engineering and Safety Section, Department of Chemical Engineering, Celestijnenlaan 200F, 3001 Leuven, Belgium
| | - Joachim Koepff
- ¶Forschungszentrum Jülich GmbH, Institute of Bio- and Geosciences, IBG-1: Biotechnology, Leo-Brandt-Straβe, 52428, Jülich, Germany
| | - Tobias Busche
- ‖Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
- **Institute for Biology-Microbiology, Freie Universität Berlin, Berlin, Germany
| | - Christian Rückert
- ‖Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Marco Oldiges
- ¶Forschungszentrum Jülich GmbH, Institute of Bio- and Geosciences, IBG-1: Biotechnology, Leo-Brandt-Straβe, 52428, Jülich, Germany
| | - Jörn Kalinowski
- ‖Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Jozef Anné
- From the ‡KU Leuven, Rega Institute, Dpt of Microbiology and Immunology, Herestraat 49, B-3000 Leuven, Belgium
| | - Jan Kormanec
- §§Institute of Molecular Biology, Slovak Academy of Sciences, Dubravska cesta 21, 84551 Bratislava, Slovakia
| | - Kristel Bernaerts
- §KU Leuven, Bio- & chemical systems Technology, Reactor Engineering and Safety Section, Department of Chemical Engineering, Celestijnenlaan 200F, 3001 Leuven, Belgium
| | - Spyridoula Karamanou
- From the ‡KU Leuven, Rega Institute, Dpt of Microbiology and Immunology, Herestraat 49, B-3000 Leuven, Belgium
| | - Anastassios Economou
- From the ‡KU Leuven, Rega Institute, Dpt of Microbiology and Immunology, Herestraat 49, B-3000 Leuven, Belgium;
| |
Collapse
|
315
|
Bürmann F, Lee BG, Than T, Sinn L, O'Reilly FJ, Yatskevich S, Rappsilber J, Hu B, Nasmyth K, Löwe J. A folded conformation of MukBEF and cohesin. Nat Struct Mol Biol 2019; 26:227-236. [PMID: 30833788 PMCID: PMC6433275 DOI: 10.1038/s41594-019-0196-z] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 01/25/2019] [Indexed: 11/09/2022]
Abstract
Structural maintenance of chromosomes (SMC)-kleisin complexes organize chromosomal DNAs in all domains of life, with key roles in chromosome segregation, DNA repair and regulation of gene expression. They function through the entrapment and active translocation of DNA, but the underlying conformational changes are largely unclear. Using structural biology, mass spectrometry and cross-linking, we investigated the architecture of two evolutionarily distant SMC-kleisin complexes: MukBEF from Escherichia coli, and cohesin from Saccharomyces cerevisiae. We show that both contain a dynamic coiled-coil discontinuity, the elbow, near the middle of their arms that permits a folded conformation. Bending at the elbow brings into proximity the hinge dimerization domain and the head-kleisin module, situated at opposite ends of the arms. Our findings favour SMC activity models that include a large conformational change in the arms, such as a relative movement between DNA contact sites during DNA loading and translocation.
Collapse
Affiliation(s)
| | | | - Thane Than
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - Ludwig Sinn
- Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Francis J O'Reilly
- Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Stanislau Yatskevich
- Department of Biochemistry, University of Oxford, Oxford, UK
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Juri Rappsilber
- Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Bin Hu
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - Kim Nasmyth
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Jan Löwe
- MRC Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
316
|
Piroli GG, Manuel AM, Patel T, Walla MD, Shi L, Lanci SA, Wang J, Galloway A, Ortinski PI, Smith DS, Frizzell N. Identification of Novel Protein Targets of Dimethyl Fumarate Modification in Neurons and Astrocytes Reveals Actions Independent of Nrf2 Stabilization. Mol Cell Proteomics 2019; 18:504-519. [PMID: 30587509 PMCID: PMC6398201 DOI: 10.1074/mcp.ra118.000922] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/01/2018] [Indexed: 12/15/2022] Open
Abstract
The fumarate ester dimethyl fumarate (DMF) has been introduced recently as a treatment for relapsing remitting multiple sclerosis (RRMS), a chronic inflammatory condition that results in neuronal demyelination and axonal loss. DMF is known to act by depleting intracellular glutathione and modifying thiols on Keap1 protein, resulting in the stabilization of the transcription factor Nrf2, which in turn induces the expression of antioxidant response element genes. We have previously shown that DMF reacts with a wide range of protein thiols, suggesting that the complete mechanisms of action of DMF are unknown. Here, we investigated other intracellular thiol residues that may also be irreversibly modified by DMF in neurons and astrocytes. Using mass spectrometry, we identified 24 novel proteins that were modified by DMF in neurons and astrocytes, including cofilin-1, tubulin and collapsin response mediator protein 2 (CRMP2). Using an in vitro functional assay, we demonstrated that DMF-modified cofilin-1 loses its activity and generates less monomeric actin, potentially inhibiting its cytoskeletal remodeling activity, which could be beneficial in the modulation of myelination during RRMS. DMF modification of tubulin did not significantly impact axonal lysosomal trafficking. We found that the oxygen consumption rate of N1E-115 neurons and the levels of proteins related to mitochondrial energy production were only slightly affected by the highest doses of DMF, confirming that DMF treatment does not impair cellular respiratory function. In summary, our work provides new insights into the mechanisms supporting the neuroprotective and remyelination benefits associated with DMF treatment in addition to the antioxidant response by Nrf2.
Collapse
Affiliation(s)
- Gerardo G Piroli
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Allison M Manuel
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Tulsi Patel
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Michael D Walla
- §Mass Spectrometry Center, Department of Chemistry & Biochemistry, University of South Carolina, Columbia, South Carolina 29205
| | - Liang Shi
- ¶Department of Biological Sciences, University of South Carolina, Columbia, South Carolina 29205
| | - Scott A Lanci
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Jingtian Wang
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Ashley Galloway
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Pavel I Ortinski
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Deanna S Smith
- ¶Department of Biological Sciences, University of South Carolina, Columbia, South Carolina 29205
| | - Norma Frizzell
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209;
| |
Collapse
|
317
|
Hörner M, Eble J, Yousefi OS, Schwarz J, Warscheid B, Weber W, Schamel WWA. Light-Controlled Affinity Purification of Protein Complexes Exemplified by the Resting ZAP70 Interactome. Front Immunol 2019; 10:226. [PMID: 30863395 PMCID: PMC6399385 DOI: 10.3389/fimmu.2019.00226] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/28/2019] [Indexed: 12/14/2022] Open
Abstract
Multiprotein complexes control the behavior of cells, such as of lymphocytes of the immune system. Methods to affinity purify protein complexes and to determine their interactome by mass spectrometry are thus widely used. One drawback of these methods is the presence of false positives. In fact, the elution of the protein of interest (POI) is achieved by changing the biochemical properties of the buffer, so that unspecifically bound proteins (the false positives) may also elute. Here, we developed an optogenetics-derived and light-controlled affinity purification method based on the light-regulated reversible protein interaction between phytochrome B (PhyB) and its phytochrome interacting factor 6 (PIF6). We engineered a truncated variant of PIF6 comprising only 22 amino acids that can be genetically fused to the POI as an affinity tag. Thereby the POI can be purified with PhyB-functionalized resin material using 660 nm light for binding and washing, and 740 nm light for elution. Far-red light-induced elution is effective but very mild as the same buffer is used for the wash and elution. As proof-of-concept, we expressed PIF-tagged variants of the tyrosine kinase ZAP70 in ZAP70-deficient Jurkat T cells, purified ZAP70 and associating proteins using our light-controlled system, and identified the interaction partners by quantitative mass spectrometry. Using unstimulated T cells, we were able to detect the known interaction partners, and could filter out all other proteins.
Collapse
Affiliation(s)
- Maximilian Hörner
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Julian Eble
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - O Sascha Yousefi
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Jennifer Schwarz
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Bettina Warscheid
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Wilfried Weber
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Wolfgang W A Schamel
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany.,Centre for Chronic Immunodeficiency CCI, Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
318
|
The classical NLRP3 inflammasome controls FADD unconventional secretion through microvesicle shedding. Cell Death Dis 2019; 10:190. [PMID: 30804327 PMCID: PMC6389912 DOI: 10.1038/s41419-019-1412-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/11/2019] [Accepted: 01/22/2019] [Indexed: 12/20/2022]
Abstract
Fas-associated death domain (FADD) is a key adaptor molecule involved in numerous physiological processes including cell death, proliferation, innate immunity and inflammation. Therefore, changes in FADD expression have dramatic cellular consequences. In mice and humans, FADD regulation can occur through protein secretion. However, the molecular mechanisms accounting for human FADD secretion were still unknown. Here we report that canonical, non-canonical, but not alternative, NLRP3 inflammasome activation in human monocytes/macrophages induced FADD secretion. NLRP3 inflammasome activation by the bacterial toxin nigericin led to the proinflammatory interleukin-1β (IL-1β) release and to the induction of cell death by pyroptosis. However, we showed that FADD secretion could occur in absence of increased IL-1β release and pyroptosis and, reciprocally, that IL-1β release and pyroptosis could occur in absence of FADD secretion. Especially, FADD, but not IL-1β, secretion following NLRP3 inflammasome activation required extracellular glucose. Thus, FADD secretion was an active process distinct from unspecific release of proteins during pyroptosis. This FADD secretion process required K+ efflux, NLRP3 sensor, ASC adaptor and CASPASE-1 molecule. Moreover, we identified FADD as a leaderless protein unconventionally secreted through microvesicle shedding, but not exosome release. Finally, we established human soluble FADD as a new marker of joint inflammation in gout and rheumatoid arthritis, two rheumatic diseases involving the NLRP3 inflammasome. Whether soluble FADD could be an actor in these diseases remains to be determined. Nevertheless, our results advance our understanding of the mechanisms contributing to the regulation of the FADD protein expression in human cells.
Collapse
|
319
|
Klein K, Sonnabend MS, Frank L, Leibiger K, Franz-Wachtel M, Macek B, Trunk T, Leo JC, Autenrieth IB, Schütz M, Bohn E. Deprivation of the Periplasmic Chaperone SurA Reduces Virulence and Restores Antibiotic Susceptibility of Multidrug-Resistant Pseudomonas aeruginosa. Front Microbiol 2019; 10:100. [PMID: 30846971 PMCID: PMC6394205 DOI: 10.3389/fmicb.2019.00100] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/17/2019] [Indexed: 12/28/2022] Open
Abstract
Pseudomonas aeruginosa is one of the main causative agents of nosocomial infections and the spread of multidrug-resistant strains is rising. Therefore, novel strategies for therapy are urgently required. The outer membrane composition of Gram-negative pathogens and especially of Pa restricts the efficacy of antibiotic entry into the cell and determines virulence. For efficient outer membrane protein biogenesis, the β-barrel assembly machinery (BAM) complex in the outer membrane and periplasmic chaperones like Skp and SurA are crucial. Previous studies indicated that the importance of individual proteins involved in outer membrane protein biogenesis may vary between different Gram-negative species. In addition, since multidrug-resistant Pa strains pose a serious global threat, the interference with both virulence and antibiotic resistance by disturbing outer membrane protein biogenesis might be a new strategy to cope with this challenge. Therefore, deletion mutants of the non-essential BAM complex components bamB and bamC, of the skp homolog hlpA as well as a conditional mutant of surA were investigated. The most profound effects for both traits were associated with reduced levels of SurA, characterized by increased membrane permeability, enhanced sensitivity to antibiotic treatment and attenuation of virulence in a Galleria mellonella infection model. Strikingly, the depletion of SurA in a multidrug-resistant clinical bloodstream isolate re-sensitized the strain to antibiotic treatment. From our data we conclude that SurA of Pa serves as a promising target for developing a drug that shows antiinfective activity and re-sensitizes multidrug-resistant strains to antibiotics.
Collapse
Affiliation(s)
- Kristina Klein
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Institut für Medizinische Mikrobiologie und Hygiene, Universität Tübingen, Tübingen, Germany
| | - Michael S. Sonnabend
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Institut für Medizinische Mikrobiologie und Hygiene, Universität Tübingen, Tübingen, Germany
| | - Lisa Frank
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Institut für Medizinische Mikrobiologie und Hygiene, Universität Tübingen, Tübingen, Germany
| | - Karolin Leibiger
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Institut für Medizinische Mikrobiologie und Hygiene, Universität Tübingen, Tübingen, Germany
| | | | - Boris Macek
- Proteome Center Tübingen, Universität Tübingen, Tübingen, Germany
| | - Thomas Trunk
- Section for Genetics and Evolutionary Biology, Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jack C. Leo
- Section for Genetics and Evolutionary Biology, Department of Biosciences, University of Oslo, Oslo, Norway
| | - Ingo B. Autenrieth
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Institut für Medizinische Mikrobiologie und Hygiene, Universität Tübingen, Tübingen, Germany
| | - Monika Schütz
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Institut für Medizinische Mikrobiologie und Hygiene, Universität Tübingen, Tübingen, Germany
| | - Erwin Bohn
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Institut für Medizinische Mikrobiologie und Hygiene, Universität Tübingen, Tübingen, Germany
| |
Collapse
|
320
|
Alves Feliciano C, Douché T, Giai Gianetto Q, Matondo M, Martin-Verstraete I, Dupuy B. CotL, a new morphogenetic spore coat protein of Clostridium difficile. Environ Microbiol 2019; 21:984-1003. [PMID: 30556639 DOI: 10.1111/1462-2920.14505] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/07/2018] [Accepted: 12/13/2018] [Indexed: 01/01/2023]
Abstract
The strict anaerobe Clostridium difficile is the most common cause of antibiotic-associated diarrhoea. The oxygen-resistant C. difficile spores play a central role in the infectious cycle, contributing to transmission, infection and recurrence. The spore surface layers, the coat and exosporium, enable the spores to resist physical and chemical stress. However, little is known about the mechanisms of their assembly. In this study, we characterized a new spore protein, CotL, which is required for the assembly of the spore coat. The cotL gene was expressed in the mother cell compartment under the dual control of the RNA polymerase sigma factors, σE and σK . CotL was localized in the spore coat, and the spores of the cotL mutant had a major morphologic defect at the level of the coat/exosporium layers. Therefore, the mutant spores contained a reduced amount of several coat/exosporium proteins and a defect in their localization in sporulating cells. Finally, cotL mutant spores were more sensitive to lysozyme and were impaired in germination, a phenotype likely to be associated with the structurally altered coat. Collectively, these results strongly suggest that CotL is a morphogenetic protein essential for the assembly of the spore coat in C. difficile.
Collapse
Affiliation(s)
- Carolina Alves Feliciano
- Laboratoire Pathogenèse des Bactéries Anaérobies, Institut Pasteur, Paris, France.,University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Thibaut Douché
- Plateforme Protéomique, Unité de Spectrométrie de Masse pour La Biologie, CNRS USR 2000, Institut Pasteur, Paris, France
| | - Quentin Giai Gianetto
- Plateforme Protéomique, Unité de Spectrométrie de Masse pour La Biologie, CNRS USR 2000, Institut Pasteur, Paris, France.,Bioinformatics and Biostatistics HUB, C3BI, CNRS USR 3756, Institut Pasteur, Paris, France
| | - Mariette Matondo
- Plateforme Protéomique, Unité de Spectrométrie de Masse pour La Biologie, CNRS USR 2000, Institut Pasteur, Paris, France
| | - Isabelle Martin-Verstraete
- Laboratoire Pathogenèse des Bactéries Anaérobies, Institut Pasteur, Paris, France.,University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Bruno Dupuy
- Laboratoire Pathogenèse des Bactéries Anaérobies, Institut Pasteur, Paris, France.,University Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
321
|
Ali M, Khan SY, Vasanth S, Ahmed MR, Chen R, Na CH, Thomson JJ, Qiu C, Gottsch JD, Riazuddin SA. Generation and Proteome Profiling of PBMC-Originated, iPSC-Derived Corneal Endothelial Cells. Invest Ophthalmol Vis Sci 2019; 59:2437-2444. [PMID: 29847650 PMCID: PMC5957521 DOI: 10.1167/iovs.17-22927] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Corneal endothelial cells (CECs) are critical in maintaining clarity of the cornea. This study was initiated to develop peripheral blood mononuclear cell (PBMC)-originated, induced pluripotent stem cell (iPSC)-derived CECs. Methods We isolated PBMCs and programmed the mononuclear cells to generate iPSCs, which were differentiated to CECs through the neural crest cells (NCCs). The morphology of differentiating iPSCs was examined at regular intervals by phase contrast microscopy. In parallel, the expression of pluripotent and corneal endothelium (CE)-associated markers was investigated by quantitative real-time PCR (qRT-PCR). The molecular architecture of the iPSC-derived CECs and human corneal endothelium (hCE) was examined by mass spectrometry–based proteome sequencing. Results The PBMC-originated, iPSC-derived CECs were tightly adherent, exhibiting a hexagonal-like shape, one of the cardinal characteristics of CECs. The CE-associated markers expressed at significantly higher levels in iPSC-derived CECs at days 13, 20, and 30 compared with their respective levels in iPSCs. It is of importance that only residual expression levels of pluripotency markers were detected in iPSC-derived CECs. Cryopreservation of iPSC-derived CECs did not affect the tight adherence of CECs and their hexagonal-like shape while expressing high levels of CE-associated markers. Mass spectrometry–based proteome sequencing identified 10,575 proteins in the iPSC-derived CEC proteome. In parallel, we completed proteome profiling of the hCE identifying 6345 proteins. Of these, 5763 proteins were identified in the iPSC-derived CECs, suggesting that 90.82% of the hCE proteome overlaps with the iPSC-derived CEC proteome. Conclusions We have successfully developed a personalized approach to generate CECs that closely mimic the molecular architecture of the hCE. To the best of our knowledge, this is the first report describing the development of PBMC-originated, iPSC-derived CECs.
Collapse
Affiliation(s)
- Muhammad Ali
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Shahid Y Khan
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Shivakumar Vasanth
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Mariya R Ahmed
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Ruiqiang Chen
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Chan Hyun Na
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Jason J Thomson
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Caihong Qiu
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, United States
| | - John D Gottsch
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - S Amer Riazuddin
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
322
|
Kesten C, Wallmann A, Schneider R, McFarlane HE, Diehl A, Khan GA, van Rossum BJ, Lampugnani ER, Szymanski WG, Cremer N, Schmieder P, Ford KL, Seiter F, Heazlewood JL, Sanchez-Rodriguez C, Oschkinat H, Persson S. The companion of cellulose synthase 1 confers salt tolerance through a Tau-like mechanism in plants. Nat Commun 2019; 10:857. [PMID: 30787279 PMCID: PMC6382854 DOI: 10.1038/s41467-019-08780-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/25/2019] [Indexed: 12/22/2022] Open
Abstract
Microtubules are filamentous structures necessary for cell division, motility and morphology, with dynamics critically regulated by microtubule-associated proteins (MAPs). Here we outline the molecular mechanism by which the MAP, COMPANION OF CELLULOSE SYNTHASE1 (CC1), controls microtubule bundling and dynamics to sustain plant growth under salt stress. CC1 contains an intrinsically disordered N-terminus that links microtubules at evenly distributed points through four conserved hydrophobic regions. By NMR and live cell analyses we reveal that two neighboring residues in the first hydrophobic binding motif are crucial for the microtubule interaction. The microtubule-binding mechanism of CC1 is reminiscent to that of the prominent neuropathology-related protein Tau, indicating evolutionary convergence of MAP functions across animal and plant cells.
Collapse
Affiliation(s)
- Christopher Kesten
- Department of Biology, ETH Zurich, 8092, Zurich, Switzerland
- School of Biosciences, University of Melbourne, Parkville, 3010, Victoria, Australia
- Max-Planck-Institute of Molecular Plant Physiology, Am Mühlenberg 1, 14476, Potsdam-Golm, Germany
| | - Arndt Wallmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), NMR-supported Structural Biology, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - René Schneider
- School of Biosciences, University of Melbourne, Parkville, 3010, Victoria, Australia
- Max-Planck-Institute of Molecular Plant Physiology, Am Mühlenberg 1, 14476, Potsdam-Golm, Germany
| | - Heather E McFarlane
- School of Biosciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Anne Diehl
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), NMR-supported Structural Biology, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Ghazanfar Abbas Khan
- School of Biosciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Barth-Jan van Rossum
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), NMR-supported Structural Biology, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Edwin R Lampugnani
- School of Biosciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Witold G Szymanski
- Max-Planck-Institute of Molecular Plant Physiology, Am Mühlenberg 1, 14476, Potsdam-Golm, Germany
| | - Nils Cremer
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), NMR-supported Structural Biology, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Peter Schmieder
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), NMR-supported Structural Biology, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Kristina L Ford
- School of Biosciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Florian Seiter
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), NMR-supported Structural Biology, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Joshua L Heazlewood
- School of Biosciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | | | - Hartmut Oschkinat
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), NMR-supported Structural Biology, Robert-Rössle-Str. 10, 13125, Berlin, Germany.
| | - Staffan Persson
- School of Biosciences, University of Melbourne, Parkville, 3010, Victoria, Australia.
- Max-Planck-Institute of Molecular Plant Physiology, Am Mühlenberg 1, 14476, Potsdam-Golm, Germany.
| |
Collapse
|
323
|
Sievers S, Metzendorf NG, Dittmann S, Troitzsch D, Gast V, Tröger SM, Wolff C, Zühlke D, Hirschfeld C, Schlüter R, Riedel K. Differential View on the Bile Acid Stress Response of Clostridioides difficile. Front Microbiol 2019; 10:258. [PMID: 30833939 PMCID: PMC6387971 DOI: 10.3389/fmicb.2019.00258] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/31/2019] [Indexed: 12/16/2022] Open
Abstract
Clostridioides difficile is an intestinal human pathogen that uses the opportunity of a depleted microbiota to cause an infection. It is known, that the composition of the intestinal bile acid cocktail has a great impact on the susceptibility toward a C. difficile infection. However, the specific response of growing C. difficile cells to diverse bile acids on the molecular level has not been described yet. In this study, we recorded proteome signatures of shock and long-term (LT) stress with the four main bile acids cholic acid (CA), chenodeoxycholic acid (CDCA), deoxycholic acid (DCA), and lithocholic acid (LCA). A general overlapping response to all tested bile acids could be determined particularly in shock experiments which appears plausible in the light of their common steroid structure. However, during LT stress several proteins showed an altered abundance in the presence of only a single or a few of the bile acids indicating the existence of specific adaptation mechanisms. Our results point at a differential induction of the groEL and dnaKJgrpE chaperone systems, both belonging to the class I heat shock genes. Additionally, central metabolic pathways involving butyrate fermentation and the reductive Stickland fermentation of leucine were effected, although CA caused a proteome signature different from the other three bile acids. Furthermore, quantitative proteomics revealed a loss of flagellar proteins in LT stress with LCA. The absence of flagella could be substantiated by electron microscopy which also indicated less flagellated cells in the presence of DCA and CDCA and no influence on flagella formation by CA. Our data break down the bile acid stress response of C. difficile into a general and a specific adaptation. The latter cannot simply be divided into a response to primary and secondary bile acids, but rather reflects a complex and variable adaptation process enabling C. difficile to survive and to cause an infection in the intestinal tract.
Collapse
Affiliation(s)
- Susanne Sievers
- Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Nicole G Metzendorf
- Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Silvia Dittmann
- Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Daniel Troitzsch
- Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Viola Gast
- Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Sophie Marlen Tröger
- Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Christian Wolff
- Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Daniela Zühlke
- Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Claudia Hirschfeld
- Department of Microbial Proteomics, Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Rabea Schlüter
- Imaging Center of the Department of Biology, University of Greifswald, Greifswald, Germany
| | - Katharina Riedel
- Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| |
Collapse
|
324
|
Wang D, Eraslan B, Wieland T, Hallström B, Hopf T, Zolg DP, Zecha J, Asplund A, Li LH, Meng C, Frejno M, Schmidt T, Schnatbaum K, Wilhelm M, Ponten F, Uhlen M, Gagneur J, Hahne H, Kuster B. A deep proteome and transcriptome abundance atlas of 29 healthy human tissues. Mol Syst Biol 2019; 15:e8503. [PMID: 30777892 PMCID: PMC6379049 DOI: 10.15252/msb.20188503] [Citation(s) in RCA: 480] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 01/01/2019] [Accepted: 01/08/2019] [Indexed: 11/28/2022] Open
Abstract
Genome-, transcriptome- and proteome-wide measurements provide insights into how biological systems are regulated. However, fundamental aspects relating to which human proteins exist, where they are expressed and in which quantities are not fully understood. Therefore, we generated a quantitative proteome and transcriptome abundance atlas of 29 paired healthy human tissues from the Human Protein Atlas project representing human genes by 18,072 transcripts and 13,640 proteins including 37 without prior protein-level evidence. The analysis revealed that hundreds of proteins, particularly in testis, could not be detected even for highly expressed mRNAs, that few proteins show tissue-specific expression, that strong differences between mRNA and protein quantities within and across tissues exist and that protein expression is often more stable across tissues than that of transcripts. Only 238 of 9,848 amino acid variants found by exome sequencing could be confidently detected at the protein level showing that proteogenomics remains challenging, needs better computational methods and requires rigorous validation. Many uses of this resource can be envisaged including the study of gene/protein expression regulation and biomarker specificity evaluation.
Collapse
Affiliation(s)
- Dongxue Wang
- Chair of Proteomics and Bioanalytics, Technische Universität München, Freising, Germany
| | - Basak Eraslan
- Computational Biology, Department of Informatics, Technical University of Munich, Garching bei München, Germany
- Department of Biochemistry, Quantitative Biosciences Munich, Gene Center, Ludwig Maximilian Universität, München, Germany
| | | | - Björn Hallström
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | | | - Daniel Paul Zolg
- Chair of Proteomics and Bioanalytics, Technische Universität München, Freising, Germany
| | - Jana Zecha
- Chair of Proteomics and Bioanalytics, Technische Universität München, Freising, Germany
| | - Anna Asplund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Li-Hua Li
- Chair of Proteomics and Bioanalytics, Technische Universität München, Freising, Germany
| | - Chen Meng
- Chair of Proteomics and Bioanalytics, Technische Universität München, Freising, Germany
| | - Martin Frejno
- Chair of Proteomics and Bioanalytics, Technische Universität München, Freising, Germany
| | - Tobias Schmidt
- Chair of Proteomics and Bioanalytics, Technische Universität München, Freising, Germany
| | | | - Mathias Wilhelm
- Chair of Proteomics and Bioanalytics, Technische Universität München, Freising, Germany
| | - Frederik Ponten
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Julien Gagneur
- Computational Biology, Department of Informatics, Technical University of Munich, Garching bei München, Germany
| | | | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technische Universität München, Freising, Germany
- Center for Integrated Protein Science Munich (CIPSM), Munich, Germany
| |
Collapse
|
325
|
Elinger D, Gabashvili A, Levin Y. Suspension Trapping (S-Trap) Is Compatible with Typical Protein Extraction Buffers and Detergents for Bottom-Up Proteomics. J Proteome Res 2019; 18:1441-1445. [DOI: 10.1021/acs.jproteome.8b00891] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Dalia Elinger
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alexandra Gabashvili
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yishai Levin
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
326
|
Gomig THB, Cavalli IJ, Souza RLRD, Vieira E, Lucena ACR, Batista M, Machado KC, Marchini FK, Marchi FA, Lima RS, de Andrade Urban C, Cavalli LR, Ribeiro EMDSF. Quantitative label-free mass spectrometry using contralateral and adjacent breast tissues reveal differentially expressed proteins and their predicted impacts on pathways and cellular functions in breast cancer. J Proteomics 2019; 199:1-14. [PMID: 30772490 DOI: 10.1016/j.jprot.2019.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/27/2019] [Accepted: 02/11/2019] [Indexed: 02/08/2023]
Abstract
Proteins play an essential role in the biological processes associated with cancer. Their altered expression levels can deregulate critical cellular pathways and interactive networks. In this study, the mass spectrometry-based label-free quantification followed by functional annotation was performed to investigate the most significant deregulated proteins among tissues of primary breast tumor (PT) and axillary metastatic lymph node (LN) and corresponding non-tumor tissues contralateral (NCT) and adjacent (ANT) from patients diagnosed with invasive ductal carcinoma. A total of 462 proteins was observed as differentially expressed (DEPs) among the groups analyzed. A high level of similarity was observed in the proteome profile of both non-tumor breast tissues and DEPs (n = 12) were mainly predicted in the RNA metabolism. The DEPs among the malignant and non-tumor breast tissues [n = 396 (PTxNCT) and n = 410 (LNxNCT)] were related to pathways of the LXR/RXR, NO, eNOS, eIF2 and sirtuins, tumor-related functions, fatty acid metabolism and oxidative stress. Remarkable similarity was observed between both malignant tissues, which the DEPs were related to metastatic capabilities. Altogether, our findings revealed differential proteomic profiles that affected cancer associated and interconnected signaling processes. Validation studies are recommended to demonstrate the potential of individual proteins and/or pathways as biological markers in breast cancer. SIGNIFICANCE: The proteomic analysis of this study revealed high similarity in the proteomic profile of the contralateral and adjacent non-tumor breast tissues. Significant differences were identified among the proteome of the malignant and non-tumor tissue groups of the same patients, providing relevant insights into the hallmarks, signaling pathways, biological functions, and interactive protein networks that act during tumorigenesis and breast cancer progression. These proteins are suggested as targets of relevant interest to be explored as potential biological markers related to tumor development and metastatic progression in the breast cancer disease.
Collapse
Affiliation(s)
| | | | | | - Evelyn Vieira
- Genetics Department, Federal University of Parana, Curitiba, Brazil
| | | | - Michel Batista
- Functional Genomics Laboratory, Carlos Chagas Institute, Fiocruz, Curitiba, Parana, Brazil; Mass Spectrometry Facility - RPT02H, Carlos Chagas Institute, Fiocruz, Curitiba, Parana, Brazil
| | | | - Fabricio Klerynton Marchini
- Functional Genomics Laboratory, Carlos Chagas Institute, Fiocruz, Curitiba, Parana, Brazil; Mass Spectrometry Facility - RPT02H, Carlos Chagas Institute, Fiocruz, Curitiba, Parana, Brazil
| | | | | | | | - Luciane Regina Cavalli
- Research Institute Pele Pequeno Principe, Curitiba, Brazil; Lombardi Comprehensive Cancer Center, Georgetown University, USA
| | | |
Collapse
|
327
|
Abstract
INTRODUCTION Small open reading frames (sORFs) with potential protein-coding capacity have been disclosed in various transcripts, including long noncoding RNAs (LncRNAs), mRNAs (5'-upstream, coding domain, and 3'-downstream), circular RNAs, pri-miRNAs, and ribosomal RNAs (rRNAs). Recent characterization of several sORF-encoded peptides (SEPs or micropeptides) revealed their important roles in many fundamental biological processes in a broad range of species from yeast to human. The success in the mining of micropeptides attributes to the advanced bioinformatics and high-throughput sequencing techniques. Areas covered: sORFs and SEPs were overlooked for their tiny size and the difficulty of identification by bioinformatics analyses. With more and more sORFs and SEPs have been identified, this field has attracted more attention. This review covers recent advances in the strategies for the detection and identification of sORFs and SEPs. Expert commentary: The advantages and drawbacks of the strategies for detection and identification of sORFs and SEPs are discussed, as well as the techniques that are used to decipher the roles of micropeptides in organisms are described.
Collapse
Affiliation(s)
- Xinqiang Yin
- a The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province , China Pharmaceutical University , Nanjing , China.,b The Basic Medical School , North Sichuan Medical College , Nanchong , China
| | - Yuanyuan Jing
- c Department of Preventive Medicine , North Sichuan Medical College , Nanchong , China
| | - Hanmei Xu
- a The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province , China Pharmaceutical University , Nanjing , China.,d State Key Laboratory of Natural Medicines, Ministry of Education , China Pharmaceutical University , Nanjing , China
| |
Collapse
|
328
|
The Foliar Endophyte Phialocephala scopiformis DAOMC 229536 Proteome When Grown on Wood Used as the Sole Carbon Source. Microbiol Resour Announc 2019; 8:MRA01280-18. [PMID: 30746512 PMCID: PMC6368647 DOI: 10.1128/mra.01280-18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/04/2019] [Indexed: 12/03/2022] Open
Abstract
The conifer needle endophyte Phialocephala scopiformis DAOMC 229536 was cultivated in medium containing ground Pinus contorta wood as the sole carbon source. Mass spectrometry analyses identified 590 proteins. The conifer needle endophyte Phialocephala scopiformis DAOMC 229536 was cultivated in medium containing ground Pinus contorta wood as the sole carbon source. Mass spectrometry analyses identified 590 proteins. The expression of extracellular hydrolases and oxidoreductases indicates a capacity to degrade wood. The results clearly demonstrate the latent saprophytic potential of P. scopiformis.
Collapse
|
329
|
Siau A, Huang X, Loh HP, Zhang N, Meng W, Sze SK, Renia L, Preiser P. Immunomic Identification of Malaria Antigens Associated With Protection in Mice. Mol Cell Proteomics 2019; 18:837-853. [PMID: 30718293 DOI: 10.1074/mcp.ra118.000997] [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: 07/30/2018] [Revised: 01/22/2019] [Indexed: 11/06/2022] Open
Abstract
Efforts to develop vaccines against malaria represent a major research target. The observations that 1) sterile protection can be obtained when the host is exposed to live parasites and 2) the immunity against blood stage parasite is principally mediated by protective antibodies suggest that a protective vaccine is feasible. However, only a small number of proteins have been investigated so far and most of the Plasmodium proteome has yet to be explored. To date, only few immunodominant antigens have emerged for testing in clinical trials but no formulation has led to substantial protection in humans. The nature of parasite molecules associated with protection remains elusive. Here, immunomic screening of mice immune sera with different protection efficiencies against the whole parasite proteome allowed us to identify a large repertoire of antigens validated by screening a library expressing antigens. The calculation of weighted scores reflecting the likelihood of protection of each antigen using five predictive criteria derived from immunomic and proteomic data sets, highlighted a priority list of protective antigens. Altogether, the approach sheds light on conserved antigens across Plasmodium that are amenable to targeting by the host immune system upon merozoite invasion and blood stage development. Most of these antigens have preliminary protection data but have not been widely considered as candidate for vaccine trials, opening new perspectives that overcome the limited choice of immunodominant, poorly protective vaccines currently being the focus of malaria vaccine researches.
Collapse
Affiliation(s)
- Anthony Siau
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore;.
| | - Ximei Huang
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore;; From the ‡Nanyang Technological University, School of Biological Sciences, Singapore
| | - Han Ping Loh
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore;; From the ‡Nanyang Technological University, School of Biological Sciences, Singapore
| | - Neng Zhang
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore
| | - Wei Meng
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore
| | - Siu Kwan Sze
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore
| | - Laurent Renia
- §Singapore Immunology Network (SIgN), A*STAR, Biopolis, Singapore
| | - Peter Preiser
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore;.
| |
Collapse
|
330
|
Chen C, Harst A, You W, Xu J, Ning K, Poetsch A. Proteomic study uncovers molecular principles of single-cell-level phenotypic heterogeneity in lipid storage of Nannochloropsis oceanica. BIOTECHNOLOGY FOR BIOFUELS 2019; 12:21. [PMID: 30740142 PMCID: PMC6360718 DOI: 10.1186/s13068-019-1361-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 01/25/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Nannochloropsis oceanica belongs to a large group of photoautotrophic eukaryotic organisms that play important roles in fixation and cycling of atmospheric CO2. Its capability of storing solar energy and carbon dioxide in the form of triacylglycerol (TAG) of up to 60% of total weight under nitrogen deprivation stress sparked interest in its use for biofuel production. Phenotypes varying in lipid accumulation among an N. oceanica population can be disclosed by single-cell analysis/sorting using fluorescence-activated cell sorting (FACS); yet the phenomenon of single cell heterogeneity in an algae population remains to be fully understood at the molecular level. In this study, combination of FACS and proteomics was used for identification, quantification and differentiation of these heterogeneities on the molecular level. RESULTS For N. oceanica cultivated under nitrogen deplete (-N) and replete (+N) conditions, two groups differing in lipid content were distinguished. These differentiations could be recognized on the population as well as the single-cell levels; proteomics uncovered alterations in carbon fixation and flux, photosynthetic machinery, lipid storage and turnover in the populations. Although heterogeneity patterns have been affected by nitrogen supply and cultivation conditions of the N. oceanica populations, differentiation itself seems to be very robust against these factors: cultivation under +N, -N, in shaker bottles, and in a photo-bioreactor all split into two subpopulations. Intriguingly, population heterogeneity resumed after subpopulations were separately recultivated for a second round, refuting the possible development of genetic heterogeneity in the course of sorting and cultivation. CONCLUSIONS This work illustrates for the first time the feasibility of combining FACS and (prote)-omics for mechanistic understanding of phenotypic heterogeneity in lipid-producing microalgae. Such combinatorial method can facilitate molecular breeding and design of bioprocesses.
Collapse
Affiliation(s)
- Chaoyun Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074 Hubei China
| | - Andreas Harst
- Plant Biochemistry, Ruhr University Bochum, 44801 Bochum, Germany
| | - Wuxin You
- Plant Biochemistry, Ruhr University Bochum, 44801 Bochum, Germany
- Single-Cell Center, CAS Key Laboratory of Biofuels and Shandong Key Laboratory of Energy Genetics, Qingdao Institute of BioEnergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, 266101 Shandong China
| | - Jian Xu
- Single-Cell Center, CAS Key Laboratory of Biofuels and Shandong Key Laboratory of Energy Genetics, Qingdao Institute of BioEnergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, 266101 Shandong China
| | - Kang Ning
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074 Hubei China
| | - Ansgar Poetsch
- Plant Biochemistry, Ruhr University Bochum, 44801 Bochum, Germany
- School of Biomedical and Healthcare Sciences, Plymouth University, Plymouth, PL4 8AA UK
| |
Collapse
|
331
|
Baberg F, Geyh S, Waldera-Lupa D, Stefanski A, Zilkens C, Haas R, Schroeder T, Stühler K. Secretome analysis of human bone marrow derived mesenchymal stromal cells. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2019; 1867:434-441. [PMID: 30716505 DOI: 10.1016/j.bbapap.2019.01.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/24/2019] [Accepted: 01/29/2019] [Indexed: 12/16/2022]
Abstract
As an essential cellular component of the bone marrow (BM) microenvironment mesenchymal stromal cells (MSC) play a pivotal role for the physiological regulation of hematopoiesis, in particular through the secretion of cytokines and chemokines. Mass spectrometry (MS) facilitates the identification and quantification of a large amount of secreted proteins (secretome), but can be hampered by the false-positive identification of contaminating proteins released from dead cells or derived from cell medium. To reduce the likelihood of contaminations we applied an approach combining secretome and proteome analysis to characterize the physiological secretome of BM derived human MSC. Our analysis revealed a secretome consisting of 315 proteins. Pathway analyses of these proteins revealed a high abundance of proteins related to cell growth and/or maintenance, signal transduction and cell communication thereby representing key biological functions of BM derived MSC on protein level. Within the MSC secretome we identified several cytokines and growth factors such as VEGFC, TGF-β1, TGF-β2 and GDF6 which are known to be involved in the physiological regulation of hematopoiesis. By comparing the peptide patterns of secretomes and cell lysates 17 proteins were identified as candidates for proteolytic processing. Taken together, our combined MS work-flow reduced the likelihood of contaminations and enabled us to carve out a specific overview about the composition of the secretome from human BM derived MSC. This methodological approach and the specific secretome signature of BM derived MSC may serve as basis for future comparative analyses of the interplay of MSC and HSPC in patients with hematological malignancies.
Collapse
Affiliation(s)
- Falk Baberg
- Institute of Molecular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Stefanie Geyh
- Department of Hematology, Oncology and Clinical Immunology, University of Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Daniel Waldera-Lupa
- Institute of Molecular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Anja Stefanski
- Molecular Proteomics Laboratory, Biomedical Research Center (BMFZ), Heinrich-Heine-University, Düsseldorf, Germany
| | - Christoph Zilkens
- Department of Orthopedic Surgery, University of Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Rainer Haas
- Department of Hematology, Oncology and Clinical Immunology, University of Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Thomas Schroeder
- Department of Hematology, Oncology and Clinical Immunology, University of Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory, Biomedical Research Center (BMFZ), Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
332
|
Lin Z, Wang RJ, Cheng Y, Du J, Volovych O, Han LB, Li JC, Hu Y, Lu ZY, Lu Z, Zou Z. Insights into the venom protein components of Microplitis mediator, an endoparasitoid wasp. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2019; 105:33-42. [PMID: 30602123 DOI: 10.1016/j.ibmb.2018.12.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/19/2018] [Accepted: 12/28/2018] [Indexed: 05/26/2023]
Abstract
Endoparasitoid wasps deliver a variety of maternal factors, such as venom proteins, viruses, and virus-like particles, from their venom and calyx fluid into hosts and thereby regulate the hosts' immune response, metabolism and development. The endoparasitoid, Microplitis mediator, is used as an important biological agent for controlling the devastating pest Helicoverpa armigera. In this study, using an integrated transcriptomic and proteomic analysis approach, we identified 75 putative venom proteins in M. mediator. The identified venom components were consistent with other known parasitoid wasps' venom proteins, including metalloproteases, serine protease inhibitors, and glycoside hydrolase family 18 enzymes. The metalloprotease and serpin family showed extensive gene duplications in venom apparatus. Isobaric tags for relative and absolute quantitation (iTRAQ) based quantitative proteomics revealed 521 proteins that were differentially expressed at 6 h and 24 h post-parasitism, including 10 wasp venom proteins that were released into the host hemolymph. Further analysis indicated that 511 differentially expressed proteins (DEP) from the host are primarily involved in the immune response, material metabolism, and extracellular matrix receptor interaction. Taken together, our results on parasitoid wasp venoms have the potential to enhance the application of endoparasitoid wasps for controlling insect pest.
Collapse
Affiliation(s)
- Zhe Lin
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Rui-Juan Wang
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Yang Cheng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jie Du
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Olga Volovych
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Li-Bin Han
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jian-Cheng Li
- Institute of Plant Protection of Hebei Academy of Agriculture and Forestry Sciences, Baoding, China
| | - Yang Hu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zi-Yun Lu
- Institute of Plant Protection of Hebei Academy of Agriculture and Forestry Sciences, Baoding, China
| | - Zhiqiang Lu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China.
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
333
|
Jones MK, Lu B, Chen DZ, Spivia WR, Mercado AT, Ljubimov AV, Svendsen CN, Van Eyk JE, Wang S. In Vitro and In Vivo Proteomic Comparison of Human Neural Progenitor Cell-Induced Photoreceptor Survival. Proteomics 2019; 19:e1800213. [PMID: 30515959 PMCID: PMC6422354 DOI: 10.1002/pmic.201800213] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/01/2018] [Indexed: 12/31/2022]
Abstract
Retinal degenerative diseases lead to blindness with few treatments. Various cell-based therapies are aimed to slow the progression of vision loss by preserving light-sensing photoreceptor cells. A subretinal injection of human neural progenitor cells (hNPCs) into the Royal College of Surgeons (RCS) rat model of retinal degeneration has aided in photoreceptor survival, though the mechanisms are mainly unknown. Identifying the retinal proteomic changes that occur following hNPC treatment leads to better understanding of neuroprotection. To mimic the retinal environment following hNPC injection, a co-culture system of retinas and hNPCs is developed. Less cell death occurs in RCS retinal tissue co-cultured with hNPCs than in retinas cultured alone, suggesting that hNPCs provide retinal protection in vitro. Comparison of ex vivo and in vivo retinas identifies nuclear factor (erythroid-derived 2)-like 2 (NRF2) mediated oxidative response signaling as an hNPC-induced pathway. This is the first study to compare proteomic changes following treatment with hNPCs in both an ex vivo and in vivo environment, further allowing the use of ex vivo modeling for mechanisms of retinal preservation. Elucidation of the protein changes in the retina following hNPC treatment may lead to the discovery of mechanisms of photoreceptor survival and its therapeutic for clinical applications.
Collapse
Affiliation(s)
- Melissa K. Jones
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center
| | - Bin Lu
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center
| | - Dawn Z. Chen
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles
| | - Weston R. Spivia
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center
| | - Augustus T. Mercado
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center
| | - Alexander V. Ljubimov
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles
| | - Clive N. Svendsen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center
| | - Jennifer E. Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles
| | - Shaomei Wang
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles
| |
Collapse
|
334
|
Park SK(R, Jung T, Thuy-Boun PS, Wang AY, Yates JR, Wolan DW. ComPIL 2.0: An Updated Comprehensive Metaproteomics Database. J Proteome Res 2019; 18:616-622. [PMID: 30525664 PMCID: PMC7767584 DOI: 10.1021/acs.jproteome.8b00722] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We designed a metaproteomic analysis method (ComPIL) to accommodate the ever-increasing number of sequences against which experimental shotgun proteomics spectra could be accurately and rapidly queried. Our objective was to create these large databases for the analysis of complex metasamples with unknown composition, including those derived from human, animal, and environmental microbiomes. The amount of high-throughput sequencing data has substantially increased since our original database was assembled in 2014. Here, we present a rebuild of the ComPIL libraries comprised of updated publicly disseminated sequence data as well as a modified version of the search engine ProLuCID-ComPIL optimized for querying experimental spectra. ComPIL 2.0 consists of 113 million protein records and roughly 4.8 billion unique tryptic peptide sequences and is 2.3 times the size of our original version. We searched a data set collected on a healthy human gut microbiome proteomic sample and compared the results to demonstrate that ComPIL 2.0 showed a substantial increase in the number of unique identified peptides and proteins compared to the first ComPIL version. The high confidence of protein identification and accuracy demonstrated by the use of ComPIL 2.0 may encourage the method's application for large-scale proteomic annotation of complex protein systems.
Collapse
Affiliation(s)
- Sung Kyu (Robin) Park
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Titus Jung
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Peter S. Thuy-Boun
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ana Y. Wang
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - John R. Yates
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Dennis W. Wolan
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
335
|
Valdés A, Holst BS, Lindersson S, Ramström M. Development of MS-based methods for identification and quantification of proteins altered during early pregnancy in dogs. J Proteomics 2019; 192:223-232. [DOI: 10.1016/j.jprot.2018.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/31/2018] [Accepted: 09/02/2018] [Indexed: 12/21/2022]
|
336
|
Flueck C, Drought LG, Jones A, Patel A, Perrin AJ, Walker EM, Nofal SD, Snijders AP, Blackman MJ, Baker DA. Phosphodiesterase beta is the master regulator of cAMP signalling during malaria parasite invasion. PLoS Biol 2019; 17:e3000154. [PMID: 30794532 PMCID: PMC6402698 DOI: 10.1371/journal.pbio.3000154] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 03/06/2019] [Accepted: 02/05/2019] [Indexed: 12/29/2022] Open
Abstract
Cyclic nucleotide signalling is a major regulator of malaria parasite differentiation. Phosphodiesterase (PDE) enzymes are known to control cyclic GMP (cGMP) levels in the parasite, but the mechanisms by which cyclic AMP (cAMP) is regulated remain enigmatic. Here, we demonstrate that Plasmodium falciparum phosphodiesterase β (PDEβ) hydrolyses both cAMP and cGMP and is essential for blood stage viability. Conditional gene disruption causes a profound reduction in invasion of erythrocytes and rapid death of those merozoites that invade. We show that this dual phenotype results from elevated cAMP levels and hyperactivation of the cAMP-dependent protein kinase (PKA). Phosphoproteomic analysis of PDEβ-null parasites reveals a >2-fold increase in phosphorylation at over 200 phosphosites, more than half of which conform to a PKA substrate consensus sequence. We conclude that PDEβ plays a critical role in governing correct temporal activation of PKA required for erythrocyte invasion, whilst suppressing untimely PKA activation during early intra-erythrocytic development.
Collapse
Affiliation(s)
- Christian Flueck
- Faculty of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Laura G. Drought
- Faculty of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Andrew Jones
- Protein Analysis and Proteomics Laboratory, the Francis Crick Institute, London, United Kingdom
| | - Avnish Patel
- Faculty of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Abigail J. Perrin
- Malaria Biochemistry Laboratory, the Francis Crick Institute, London, United Kingdom
| | - Eloise M. Walker
- Faculty of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Stephanie D. Nofal
- Faculty of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Ambrosius P. Snijders
- Protein Analysis and Proteomics Laboratory, the Francis Crick Institute, London, United Kingdom
| | - Michael J. Blackman
- Faculty of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Malaria Biochemistry Laboratory, the Francis Crick Institute, London, United Kingdom
| | - David A. Baker
- Faculty of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
337
|
Gaviard C, Cosette P, Jouenne T, Hardouin J. LasB and CbpD Virulence Factors of Pseudomonas aeruginosa Carry Multiple Post-Translational Modifications on Their Lysine Residues. J Proteome Res 2019; 18:923-933. [PMID: 30672296 DOI: 10.1021/acs.jproteome.8b00556] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pseudomonas aeruginosa is a multi-drug resistant human pathogen largely involved in nosocomial infections. Today, effective antibacterial agents are lacking. Exploring the bacterial physiology at the post-translational modifications (PTM) level may contribute to the renewal of fighting strategies. Indeed, some correlations between PTMs and the bacterial virulence, adaptation, and resistance have been shown. In a previous study performed in P. aeruginosa, we reported that many virulence factors like chitin-binding protein CbpD and elastase LasB were multiphosphorylated. Besides phosphorylation, other PTMs, like those occurring on lysine, seem to play key roles in bacteria. In the present study, we investigated for the first time the lysine succinylome and acetylome of the extracellular compartment of P. aeruginosa by using a two-dimensional immunoaffinity approach. Some virulence factors were identified as multimodified on lysine residues, among them, LasB and CbpD. Lysine can be modified by a wide range of chemical groups. In order to check the presence of other chemical groups on modified lysines identified on LasB and CbpD, we used 1- and 2- dimensional gel electrophoresis approaches to target lysine modified by 7 other modifications: butyrylation, crotonylation, dimethylation, malonylation, methylation, propionylation, and trimethylation. We showed that some lysines of these two virulence factors were modified by these 9 different PTMs. Interestingly, we found that the PTMs recovered on these two virulence factors were different than those previously reported in the intracellular compartment.
Collapse
Affiliation(s)
- Charlotte Gaviard
- Normandie University, UNIROUEN, INSA Rouen, CNRS, PBS , 76000 Rouen , France.,PISSARO Proteomic Facility, IRIB , 76821 Mont-Saint-Aignan , France
| | - Pascal Cosette
- Normandie University, UNIROUEN, INSA Rouen, CNRS, PBS , 76000 Rouen , France.,PISSARO Proteomic Facility, IRIB , 76821 Mont-Saint-Aignan , France
| | - Thierry Jouenne
- Normandie University, UNIROUEN, INSA Rouen, CNRS, PBS , 76000 Rouen , France.,PISSARO Proteomic Facility, IRIB , 76821 Mont-Saint-Aignan , France
| | - Julie Hardouin
- Normandie University, UNIROUEN, INSA Rouen, CNRS, PBS , 76000 Rouen , France.,PISSARO Proteomic Facility, IRIB , 76821 Mont-Saint-Aignan , France
| |
Collapse
|
338
|
Ali HR, Assiri MA, Harris PS, Michel CR, Yun Y, Marentette JO, Huynh FK, Orlicky DJ, Shearn CT, Saba LM, Reisdorph R, Reisdorph N, Hirschey MD, Fritz KS. Quantifying Competition among Mitochondrial Protein Acylation Events Induced by Ethanol Metabolism. J Proteome Res 2019; 18:1513-1531. [PMID: 30644754 DOI: 10.1021/acs.jproteome.8b00800] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mitochondrial dysfunction is one of many key factors in the etiology of alcoholic liver disease (ALD). Lysine acetylation is known to regulate numerous mitochondrial metabolic pathways, and recent reports demonstrate that alcohol-induced protein acylation negatively impacts these processes. To identify regulatory mechanisms attributed to alcohol-induced protein post-translational modifications, we employed a model of alcohol consumption within the context of wild type (WT), sirtuin 3 knockout (SIRT3 KO), and sirtuin 5 knockout (SIRT5 KO) mice to manipulate hepatic mitochondrial protein acylation. Mitochondrial fractions were examined by label-free quantitative HPLC-MS/MS to reveal competition between lysine acetylation and succinylation. A class of proteins defined as "differential acyl switching proteins" demonstrate select sensitivity to alcohol-induced protein acylation. A number of these proteins reveal saturated lysine-site occupancy, suggesting a significant level of differential stoichiometry in the setting of ethanol consumption. We hypothesize that ethanol downregulates numerous mitochondrial metabolic pathways through differential acyl switching proteins. Data are available via ProteomeXchange with identifier PXD012089.
Collapse
Affiliation(s)
- Hadi R Ali
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Mohammed A Assiri
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Peter S Harris
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Cole R Michel
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Youngho Yun
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - John O Marentette
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Frank K Huynh
- Department of Biological Sciences , San Jose State University , San Jose , California 95192 , United States
| | - David J Orlicky
- Department of Pathology, School of Medicine , University of Colorado Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Colin T Shearn
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Laura M Saba
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Richard Reisdorph
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Nichole Reisdorph
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Matthew D Hirschey
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Department of Pharmacology and Cancer Biology , Duke University Medical Center , Durham , North Carolina 27710 , United States
| | - Kristofer S Fritz
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| |
Collapse
|
339
|
Deleon-Pennell KY, Ero OK, Ma Y, Padmanabhan Iyer R, Flynn ER, Espinoza I, Musani SK, Vasan RS, Hall ME, Fox ER, Lindsey ML. Glycoproteomic Profiling Provides Candidate Myocardial Infarction Predictors of Later Progression to Heart Failure. ACS OMEGA 2019; 4:1272-1280. [PMID: 30729226 PMCID: PMC6356850 DOI: 10.1021/acsomega.8b02207] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/20/2018] [Indexed: 05/10/2023]
Abstract
We hypothesized that identifying plasma glycoproteins that predict the development of heart failure following myocardial infarction (MI) could help to stratify subjects at risk. Plasma collected at visit 2 (2005-2008) from an MI subset of Jackson Heart Study participants underwent glycoproteomics and was grouped by the outcome: (1) heart failure hospitalization after visit 2 (n = 15) and (2) without hospitalization by 2012 (n = 45). Proteins were mapped for biological processes and functional pathways using Ingenuity Pathway Analysis and linked to clinical characteristics. A total of 198 glycopeptides corresponding to 88 proteins were identified (data available via ProteomeXchange with identifier PXD009870). Of these, 14 glycopeptides were significantly different between MI and MI + HF groups and corresponded to apolipoprotein (Apo) F, transthyretin, Apo C-IV, prostaglandin-D2 synthase, complement C9, and CD59 (p < 0.05 for all). All proteins were elevated in the MI + HF group, except CD59, which was lower. Four canonical pathways were upregulated in the MI + HF group (p < 0.05 for all): acute phase response, liver X receptor/retinoid X receptor, and macrophage reactive oxygen species generation. The coagulation pathway was significantly downregulated in the MI + HF group (p < 0.05). Even after adjustment for age and sex, Apo F was associated with the increased risk for heart failure (OR = 21.84; 95% CI 3.20-149.14). In conclusion, glycoproteomic profiling provided candidate early MI predictors of later progression to heart failure.
Collapse
Affiliation(s)
- Kristine Y. Deleon-Pennell
- Mississippi
Center for Heart Research, Department of Physiology and
Biophysics, Department of Preventive Medicine and Cancer Institute, Jackson Heart Study, and Division of Cardiology, UMMC, Jackson, Mississippi 39216-4505, United States
- Research
Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, Mississippi 39216, United States
- E-mail: . Phone: 843-789-6839. Fax: 843-876-5068 (K.Y.D.-P.)
| | - Osasere K. Ero
- Mississippi
Center for Heart Research, Department of Physiology and
Biophysics, Department of Preventive Medicine and Cancer Institute, Jackson Heart Study, and Division of Cardiology, UMMC, Jackson, Mississippi 39216-4505, United States
| | - Yonggang Ma
- Mississippi
Center for Heart Research, Department of Physiology and
Biophysics, Department of Preventive Medicine and Cancer Institute, Jackson Heart Study, and Division of Cardiology, UMMC, Jackson, Mississippi 39216-4505, United States
| | - Rugmani Padmanabhan Iyer
- Mississippi
Center for Heart Research, Department of Physiology and
Biophysics, Department of Preventive Medicine and Cancer Institute, Jackson Heart Study, and Division of Cardiology, UMMC, Jackson, Mississippi 39216-4505, United States
| | - Elizabeth R. Flynn
- Mississippi
Center for Heart Research, Department of Physiology and
Biophysics, Department of Preventive Medicine and Cancer Institute, Jackson Heart Study, and Division of Cardiology, UMMC, Jackson, Mississippi 39216-4505, United States
| | - Ingrid Espinoza
- Mississippi
Center for Heart Research, Department of Physiology and
Biophysics, Department of Preventive Medicine and Cancer Institute, Jackson Heart Study, and Division of Cardiology, UMMC, Jackson, Mississippi 39216-4505, United States
| | - Solomon K. Musani
- Mississippi
Center for Heart Research, Department of Physiology and
Biophysics, Department of Preventive Medicine and Cancer Institute, Jackson Heart Study, and Division of Cardiology, UMMC, Jackson, Mississippi 39216-4505, United States
| | - Ramachandran S. Vasan
- Preventive
Medicine and Epidemiology and Cardiology, Department of Medicine, Boston University School of Medicine, Boston University, Boston, Massachusetts 02118, United States
| | - Michael E. Hall
- Mississippi
Center for Heart Research, Department of Physiology and
Biophysics, Department of Preventive Medicine and Cancer Institute, Jackson Heart Study, and Division of Cardiology, UMMC, Jackson, Mississippi 39216-4505, United States
| | - Ervin R. Fox
- Mississippi
Center for Heart Research, Department of Physiology and
Biophysics, Department of Preventive Medicine and Cancer Institute, Jackson Heart Study, and Division of Cardiology, UMMC, Jackson, Mississippi 39216-4505, United States
| | - Merry L. Lindsey
- Mississippi
Center for Heart Research, Department of Physiology and
Biophysics, Department of Preventive Medicine and Cancer Institute, Jackson Heart Study, and Division of Cardiology, UMMC, Jackson, Mississippi 39216-4505, United States
- Research
Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, Mississippi 39216, United States
- E-mail: . Phone: 601-815-1329. Fax: 601-984-1817 (M.L.L.)
| |
Collapse
|
340
|
Anjo SI, Melo MN, Loureiro LR, Sabala L, Castanheira P, Grãos M, Manadas B. oxSWATH: An integrative method for a comprehensive redox-centered analysis combined with a generic differential proteomics screening. Redox Biol 2019; 22:101130. [PMID: 30737169 PMCID: PMC6435957 DOI: 10.1016/j.redox.2019.101130] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 12/21/2022] Open
Abstract
Most of the redox proteomics strategies are focused on the identification and relative quantification of cysteine oxidation without considering the variation in the total levels of the proteins. However, protein synthesis and protein degradation also belong to the regulatory mechanisms of the cells, being therefore important to consider the changes in total protein levels in PTMs-focused analyses, such as cysteine redox characterization. Therefore, a novel integrative approach combining the SWATH-MS method with differential alkylation using a combination of commonly available alkylating reagents (oxSWATH) is presented, by which it is possible to integrate the information regarding relative cysteine oxidation with the analysis of the total protein levels in a cost-effective high-throughput approach. The proposed method was tested using a redox-regulated protein and further applied to a comparative analysis of secretomes obtained from cells cultured under control or oxidative stress conditions to strengthen the importance of considering the overall proteome changes. Using the OxSWATH method it was possible to determine both the relative proportion of reduced and reversible oxidized oxoforms, as well as the total levels of each oxoform by taking into consideration the total levels of the protein. Therefore, using OxSWATH the comparative analyses can be performed at two different levels by considering the relative proportion or the total levels at both peptide and protein level. Moreover, since samples are acquired in SWATH-MS mode, besides the redox centered analysis, a generic differential protein expression analysis can also be performed, allowing a truly comprehensive evaluation of proteomics changes upon the oxidative stimulus. Data are available via ProteomeXchange and SWATHAtlas with the identifiers PXD006802, PXD006802, and PASS01210. Determination of redox changes considering protein total levels. Integrative redoxomics and common differential proteomics in a single analysis. Differential alkylation strategy using commonly available alkylating agents. First untargeted label-free quantitative method to study cysteine oxidation.
Collapse
Affiliation(s)
- Sandra I Anjo
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| | - Matilde N Melo
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Liliana R Loureiro
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Lúcia Sabala
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | | | - Mário Grãos
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Biocant, Technology Transfer Association, Cantanhede, Portugal
| | - Bruno Manadas
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
341
|
Eukaryotic Translation Elongation is Modulated by Single Natural Nucleotide Derivatives in the Coding Sequences of mRNAs. Genes (Basel) 2019; 10:genes10020084. [PMID: 30691071 PMCID: PMC6409545 DOI: 10.3390/genes10020084] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/14/2019] [Accepted: 01/23/2019] [Indexed: 12/16/2022] Open
Abstract
RNA modifications are crucial factors for efficient protein synthesis. All classes of RNAs that are involved in translation are modified to different extents. Recently, mRNA modifications and their impact on gene regulation became a focus of interest because they can exert a variety of effects on the fate of mRNAs. mRNA modifications within coding sequences can either directly or indirectly interfere with protein synthesis. In order to investigate the roles of various natural occurring modified nucleotides, we site-specifically introduced them into the coding sequence of reporter mRNAs and subsequently translated them in HEK293T cells. The analysis of the respective protein products revealed a strong position-dependent impact of RNA modifications on translation efficiency and accuracy. Whereas a single 5-methylcytosine (m5C) or pseudouridine (Ψ) did not reduce product yields, N1-methyladenosine (m1A) generally impeded the translation of the respective modified mRNA. An inhibitory effect of 2′O-methlyated nucleotides (Nm) and N6-methyladenosine (m6A) was strongly dependent on their position within the codon. Finally, we could not attribute any miscoding potential to the set of mRNA modifications tested in HEK293T cells.
Collapse
|
342
|
Schwill M, Tamaskovic R, Gajadhar AS, Kast F, White FM, Plückthun A. Systemic analysis of tyrosine kinase signaling reveals a common adaptive response program in a HER2-positive breast cancer. Sci Signal 2019; 12:12/565/eaau2875. [PMID: 30670633 DOI: 10.1126/scisignal.aau2875] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Drug-induced compensatory signaling and subsequent rewiring of the signaling pathways that support cell proliferation and survival promote the development of acquired drug resistance in tumors. Here, we sought to analyze the adaptive kinase response in cancer cells after distinct treatment with agents targeting human epidermal growth factor receptor 2 (HER2), specifically those that induce either only temporary cell cycle arrest or, alternatively, apoptosis in HER2-overexpressing cancers. We compared trastuzumab, ARRY380, the combination thereof, and a biparatopic, HER2-targeted designed ankyrin repeat protein (DARPin; specifically, 6L1G) and quantified the phosphoproteome by isobaric tagging using tandem mass tag liquid chromatography/tandem mass spectrometry (TMT LC-MS/MS). We found a specific signature of persistently phosphorylated tyrosine peptides after the nonapoptotic treatments, which we used to distinguish between different treatment-induced cancer cell fates. Next, we analyzed the activation of serine/threonine and tyrosine kinases after treatment using a bait peptide chip array and predicted the corresponding active kinases. Through a combined system-wide analysis, we identified a common adaptive kinase response program that involved the activation of focal adhesion kinase 1 (FAK1), protein kinase C-δ (PRKCD), and Ephrin (EPH) family receptors. These findings reveal potential targets to prevent adaptive resistance to HER2-targeted therapies.
Collapse
Affiliation(s)
- Martin Schwill
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland
| | - Rastislav Tamaskovic
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland
| | - Aaron S Gajadhar
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Florian Kast
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland
| | - Forest M White
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland.
| |
Collapse
|
343
|
Regl C, Wohlschlager T, Esser-Skala W, Wagner I, Samonig M, Holzmann J, Huber CG. Dilute-and-shoot analysis of therapeutic monoclonal antibody variants in fermentation broth: a method capability study. MAbs 2019; 11:569-582. [PMID: 30668249 PMCID: PMC6512939 DOI: 10.1080/19420862.2018.1563034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Monoclonal antibodies (mAbs) are widely applied as highly specific and efficient therapeutic agents for various medical conditions, including cancer, inflammatory and autoimmune diseases. As protein production in cellular systems inherently generates a multitude of molecular variants, manufacturing of mAbs requires stringent control in order to ensure safety and efficacy of the drugs. Moreover, monitoring of mAb variants in the course of the fermentation process may allow instant tuning of process parameters to maintain optimal cell culture conditions. Here, we describe a fast and robust workflow for the characterization of mAb variants in fermentation broth. Sample preparation is minimal in that the fermentation broth is shortly centrifuged before dilution and HPLC-MS analysis in a short 15-min gradient run. In a single analysis, N-glycosylation and truncation variants of the expressed mAb are identified at the intact protein level. Simultaneously, absolute quantification of mAb content in fermentation broth is achieved. The whole workflow features excellent robustness as well as retention time and peak area stability. Additional enzymatic removal of N-glycans enables determination of mAb glycation levels, which are subsequently considered in relative N-glycoform quantification to correct for isobaric galactosylation. Several molecular attributes of the expressed therapeutic protein may thus be continuously monitored to ensure the desired product profile. Application of the described workflow in an industrial environment may therefore substantially enhance in-process control in mAb production, as well as targeted biosimilar development.
Collapse
Affiliation(s)
- Christof Regl
- a Department of Biosciences, Bioanalytical Research Labs , University of Salzburg , Salzburg , Austria.,b Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization , University of Salzburg , Salzburg , Austria
| | - Therese Wohlschlager
- a Department of Biosciences, Bioanalytical Research Labs , University of Salzburg , Salzburg , Austria.,b Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization , University of Salzburg , Salzburg , Austria
| | - Wolfgang Esser-Skala
- a Department of Biosciences, Bioanalytical Research Labs , University of Salzburg , Salzburg , Austria.,b Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization , University of Salzburg , Salzburg , Austria
| | - Iris Wagner
- a Department of Biosciences, Bioanalytical Research Labs , University of Salzburg , Salzburg , Austria.,b Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization , University of Salzburg , Salzburg , Austria
| | - Martin Samonig
- a Department of Biosciences, Bioanalytical Research Labs , University of Salzburg , Salzburg , Austria.,b Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization , University of Salzburg , Salzburg , Austria.,c Thermo Fisher Scientific GmbH , Germering , Germany
| | - Johann Holzmann
- b Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization , University of Salzburg , Salzburg , Austria.,d Technical Development Biosimilars , Global Drug Development, Novartis, Sandoz GmbH , Kundl , Austria
| | - Christian G Huber
- a Department of Biosciences, Bioanalytical Research Labs , University of Salzburg , Salzburg , Austria.,b Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization , University of Salzburg , Salzburg , Austria
| |
Collapse
|
344
|
Campagne A, Lee MK, Zielinski D, Michaud A, Le Corre S, Dingli F, Chen H, Shahidian LZ, Vassilev I, Servant N, Loew D, Pasmant E, Postel-Vinay S, Wassef M, Margueron R. BAP1 complex promotes transcription by opposing PRC1-mediated H2A ubiquitylation. Nat Commun 2019; 10:348. [PMID: 30664650 PMCID: PMC6341105 DOI: 10.1038/s41467-018-08255-x] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 12/21/2018] [Indexed: 11/09/2022] Open
Abstract
In Drosophila, a complex consisting of Calypso and ASX catalyzes H2A deubiquitination and has been reported to act as part of the Polycomb machinery in transcriptional silencing. The mammalian homologs of these proteins (BAP1 and ASXL1/2/3, respectively), are frequently mutated in various cancer types, yet their precise functions remain unclear. Using an integrative approach based on isogenic cell lines generated with CRISPR/Cas9, we uncover an unanticipated role for BAP1 in gene activation. This function requires the assembly of an enzymatically active BAP1-associated core complex (BAP1.com) containing one of the redundant ASXL proteins. We investigate the mechanism underlying BAP1.com-mediated transcriptional regulation and show that it does not participate in Polycomb-mediated silencing. Instead, our results establish that the function of BAP1.com is to safeguard transcriptionally active genes against silencing by the Polycomb Repressive Complex 1. In Drosophila, the Calypso–ASX complex catalyzes H2A deubiquitination and aids Polycomb in transcriptional silencing. Here the authors show that the orthologous complex, BAP1.com, promotes gene activation by counteracting PRC1-mediated gene silencing.
Collapse
Affiliation(s)
- Antoine Campagne
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France
| | - Ming-Kang Lee
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France
| | - Dina Zielinski
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France.,INSERM U900, Mines ParisTech, 75005, Paris, France
| | - Audrey Michaud
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France
| | - Stéphanie Le Corre
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France
| | - Florent Dingli
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France
| | - Hong Chen
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France
| | - Lara Z Shahidian
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Ivaylo Vassilev
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U934/CNRS UMR3215, 75005, Paris, France.,INSERM U900, Mines ParisTech, 75005, Paris, France
| | - Nicolas Servant
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France.,INSERM U900, Mines ParisTech, 75005, Paris, France
| | - Damarys Loew
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France
| | - Eric Pasmant
- Department of Molecular Genetics Pathology, Cochin Hospital, HUPC AP-HP, EA7331, Faculty of Pharmacy, University of Paris Descartes, Paris, 75014, France
| | - Sophie Postel-Vinay
- Département d'Innovation Thérapeutique et Essais Précoces, INSERM U981, Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, France
| | - Michel Wassef
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France. .,INSERM U934/CNRS UMR3215, 75005, Paris, France.
| | - Raphaël Margueron
- Institut Curie, Paris Sciences et Lettres Research University, Sorbonne University, 75005, Paris, France. .,INSERM U934/CNRS UMR3215, 75005, Paris, France.
| |
Collapse
|
345
|
Rice SJ, Liu X, Zhang J, Belani CP. Absolute Quantification of All Identified Plasma Proteins from SWATH Data for Biomarker Discovery. Proteomics 2019; 19:e1800135. [DOI: 10.1002/pmic.201800135] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 09/27/2018] [Indexed: 02/05/2023]
Affiliation(s)
- Shawn J. Rice
- Penn State Cancer InstitutePenn State College of Medicine Hershey PA 17033 USA
| | - Xin Liu
- Penn State Cancer InstitutePenn State College of Medicine Hershey PA 17033 USA
| | - Jianhong Zhang
- Penn State Cancer InstitutePenn State College of Medicine Hershey PA 17033 USA
| | - Chandra P. Belani
- Penn State Cancer InstitutePenn State College of Medicine Hershey PA 17033 USA
- Department of MedicinePenn State College of Medicine Hershey PA 17033 USA
| |
Collapse
|
346
|
Mycobacteria and their sweet proteins: An overview of protein glycosylation and lipoglycosylation in M. tuberculosis. Tuberculosis (Edinb) 2019; 115:1-13. [PMID: 30948163 DOI: 10.1016/j.tube.2019.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 12/16/2022]
Abstract
Post-translational modifications represent a key aspect of enzyme and protein regulation and function. Post-translational modifications are involved in signaling and response to stress, adaptation to changing environments, regulation of toxic and damaged proteins, proteins localization and host-pathogen interactions. Glycosylation in Mycobacterium tuberculosis (Mtb), is a post-translational modification often found in conjunction with acylation in mycobacterial proteins. Since the discovery of glycosylated proteins in the early 1980's, important advances in our understanding of the mechanisms of protein glycosylation have been made. The number of known glycosylated substrates in Mtb has grown through the years, yet many questions remain. This review will explore the current knowledge on protein glycosylation in Mtb, causative agent of Tuberculosis and number one infectious killer in the world. The mechanism and significance of this post-translational modification, as well as maturation, export and acylation of glycosylated proteins will be reviewed. We expect to provide the reader with an overall view of protein glycosylation in Mtb, as well as the significance of this post-translational modification to the physiology and host-pathogen interactions of this important pathogen. The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD011081 and 10.6019/PXD011081.
Collapse
|
347
|
Mouton AJ, Ma Y, Rivera Gonzalez OJ, Daseke MJ, Flynn ER, Freeman TC, Garrett MR, DeLeon-Pennell KY, Lindsey ML. Fibroblast polarization over the myocardial infarction time continuum shifts roles from inflammation to angiogenesis. Basic Res Cardiol 2019; 114:6. [PMID: 30635789 PMCID: PMC6329742 DOI: 10.1007/s00395-019-0715-4] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 01/04/2019] [Indexed: 12/11/2022]
Abstract
Cardiac fibroblasts are the major producers of extracellular matrix (ECM) to form infarct scar. We hypothesized that fibroblasts undergo a spectrum of phenotype states over the course of myocardial infarction (MI) from early onset to scar formation. Fibroblasts were isolated from the infarct region of C57BL/6J male mice (3-6 months old, n = 60) at days 0 (no MI control) and 1, 3, or 7 after MI. Whole transcriptome analysis was performed by RNA-sequencing. Of the genes sequenced, 3371 were differentially expressed after MI. Enrichment analysis revealed that MI day 1 fibroblasts displayed pro-inflammatory, leukocyte-recruiting, pro-survival, and anti-migratory phenotype through Tnfrsf9 and CD137 signaling. MI day 3 fibroblasts had a proliferative, pro-fibrotic, and pro-angiogenic profile with elevated Il4ra signaling. MI day 7 fibroblasts showed an anti-angiogenic homeostatic-like myofibroblast profile and with a step-wise increase in Acta2 expression. MI day 7 fibroblasts relied on Pik3r3 signaling to mediate Tgfb1 effects and Fgfr2 to regulate PI3K signaling. In vitro, the day 3 MI fibroblast secretome stimulated angiogenesis, while day 7 MI fibroblast secretome repressed angiogenesis through Thbs1 signaling. Our results reveal novel mechanisms for fibroblasts in expressing pro-inflammatory molecules and regulating angiogenesis following MI.
Collapse
Affiliation(s)
- Alan J Mouton
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St, Jackson, MS, 39216-4505, USA
| | - Yonggang Ma
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St, Jackson, MS, 39216-4505, USA
| | - Osvaldo J Rivera Gonzalez
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St, Jackson, MS, 39216-4505, USA
| | - Michael J Daseke
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St, Jackson, MS, 39216-4505, USA
| | - Elizabeth R Flynn
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St, Jackson, MS, 39216-4505, USA
| | - Tom C Freeman
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Michael R Garrett
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Kristine Y DeLeon-Pennell
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St, Jackson, MS, 39216-4505, USA
- Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, 39216, USA
| | - Merry L Lindsey
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 2500 North State St, Jackson, MS, 39216-4505, USA.
- Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, 39216, USA.
| |
Collapse
|
348
|
Pirro M, Schoof E, van Vliet SJ, Rombouts Y, Stella A, de Ru A, Mohammed Y, Wuhrer M, van Veelen PA, Hensbergen PJ. Glycoproteomic Analysis of MGL-Binding Proteins on Acute T-Cell Leukemia Cells. J Proteome Res 2019; 18:1125-1132. [PMID: 30582698 PMCID: PMC6399673 DOI: 10.1021/acs.jproteome.8b00796] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
![]()
C-type lectins are
a diverse group of proteins involved in many
human physiological and pathological processes. Most C-type lectins
are glycan-binding proteins, some of which are pivotal for innate
immune responses against pathogens. Other C-type lectins, such as
the macrophage galactose-type lectin (MGL), have been shown to induce
immunosuppressive responses upon the recognition of aberrant glycosylation
on cancer cells. MGL is known to recognize terminal N-acetylgalactosamine (GalNAc), such as the Tn antigen, which is commonly
found on malignant cells. Even though this glycan specificity of MGL
is well described, there is a lack of understanding of the actual
glycoproteins that bind MGL. We present a glycoproteomic workflow
for the identification of MGL-binding proteins, which we applied to
study MGL ligands on the human Jurkat leukemia cell line. In addition
to the known MGL ligands and Tn antigen-carrying proteins CD43 and
CD45 on these cells, we have identified a set of novel cell-surface
ligands for MGL. Importantly, for several of these, O-glycosylation
has hitherto not been described. Altogether, our data provide new
insight into the identification and structure of novel MGL ligands
that presumably act as modulatory molecules in cancer immune responses.
Collapse
Affiliation(s)
- Martina Pirro
- Center for Proteomics and Metabolomics , Leiden University Medical Center , 2300 RC Leiden , The Netherlands
| | - Esmee Schoof
- Center for Proteomics and Metabolomics , Leiden University Medical Center , 2300 RC Leiden , The Netherlands
| | - Sandra J van Vliet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Dept. of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection & Immunity Institute, 1007 MB Amsterdam , The Netherlands
| | - Yoann Rombouts
- Institut de Pharmacologie et de Biologie Structurale , Université de Toulouse, CNRS, UPS , Toulouse 31062 , France
| | - Alexandre Stella
- Institut de Pharmacologie et de Biologie Structurale , Université de Toulouse, CNRS, UPS , Toulouse 31062 , France
| | - Arnoud de Ru
- Center for Proteomics and Metabolomics , Leiden University Medical Center , 2300 RC Leiden , The Netherlands
| | - Yassene Mohammed
- Center for Proteomics and Metabolomics , Leiden University Medical Center , 2300 RC Leiden , The Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics , Leiden University Medical Center , 2300 RC Leiden , The Netherlands
| | - Peter A van Veelen
- Center for Proteomics and Metabolomics , Leiden University Medical Center , 2300 RC Leiden , The Netherlands
| | - Paul J Hensbergen
- Center for Proteomics and Metabolomics , Leiden University Medical Center , 2300 RC Leiden , The Netherlands
| |
Collapse
|
349
|
Selli C, Turnbull AK, Pearce DA, Li A, Fernando A, Wills J, Renshaw L, Thomas JS, Dixon JM, Sims AH. Molecular changes during extended neoadjuvant letrozole treatment of breast cancer: distinguishing acquired resistance from dormant tumours. Breast Cancer Res 2019; 21:2. [PMID: 30616553 PMCID: PMC6323855 DOI: 10.1186/s13058-018-1089-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 12/19/2018] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The risk of recurrence for endocrine-treated breast cancer patients persists for many years or even decades following surgery and apparently successful adjuvant therapy. This period of dormancy and acquired resistance is inherently difficult to investigate; previous efforts have been limited to in-vitro or in-vivo approaches. In this study, sequential tumour samples from patients receiving extended neoadjuvant aromatase inhibitor therapy were characterised as a novel clinical model. METHODS Consecutive tumour samples from 62 patients undergoing extended (4-45 months) neoadjuvant aromatase inhibitor therapy with letrozole were subjected to transcriptomic and proteomic analysis, representing before (≤ 0), early (13-120 days), and long-term (> 120 days) neoadjuvant aromatase inhibitor therapy with letrozole. Patients with at least a 40% initial reduction in tumour size by 4 months of treatment were included. Of these, 42 patients with no subsequent progression were classified as "dormant", and the remaining 20 patients as "acquired resistant". RESULTS Changes in gene expression in dormant tumours begin early and become more pronounced at later time points. Therapy-induced changes in resistant tumours were common features of treatment, rather than being specific to the resistant phenotype. Comparative analysis of long-term treated dormant and resistant tumours highlighted changes in epigenetics pathways including DNA methylation and histone acetylation. The DNA methylation marks 5-methylcytosine and 5-hydroxymethylcytosine were significantly reduced in resistant tumours compared with dormant tissues after extended letrozole treatment. CONCLUSIONS This is the first patient-matched gene expression study investigating long-term aromatase inhibitor-induced dormancy and acquired resistance in breast cancer. Dormant tumours continue to change during treatment whereas acquired resistant tumours more closely resemble their diagnostic samples. Global loss of DNA methylation was observed in resistant tumours under extended treatment. Epigenetic alterations may lead to escape from dormancy and drive acquired resistance in a subset of patients, supporting a potential role for therapy targeted at these epigenetic alterations in the management of resistance to oestrogen deprivation therapy.
Collapse
Affiliation(s)
- Cigdem Selli
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK.,Department of Pharmacology, Faculty of Pharmacy, Ege University, 35040, Izmir, Turkey
| | - Arran K Turnbull
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK.,Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - Dominic A Pearce
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK
| | - Ang Li
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK
| | - Anu Fernando
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK.,Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - Jimi Wills
- Mass Spectrometry Unit, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK
| | - Lorna Renshaw
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - Jeremy S Thomas
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - J Michael Dixon
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - Andrew H Sims
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK.
| |
Collapse
|
350
|
Chen Z, Amro EM, Becker F, Hölzer M, Rasa SMM, Njeru SN, Han B, Di Sanzo S, Chen Y, Tang D, Tao S, Haenold R, Groth M, Romanov VS, Kirkpatrick JM, Kraus JM, Kestler HA, Marz M, Ori A, Neri F, Morita Y, Rudolph KL. Cohesin-mediated NF-κB signaling limits hematopoietic stem cell self-renewal in aging and inflammation. J Exp Med 2019; 216:152-175. [PMID: 30530755 PMCID: PMC6314529 DOI: 10.1084/jem.20181505] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/06/2018] [Accepted: 11/19/2018] [Indexed: 01/02/2023] Open
Abstract
Organism aging is characterized by increased inflammation and decreased stem cell function, yet the relationship between these factors remains incompletely understood. This study shows that aged hematopoietic stem and progenitor cells (HSPCs) exhibit increased ground-stage NF-κB activity, which enhances their responsiveness to undergo differentiation and loss of self-renewal in response to inflammation. The study identifies Rad21/cohesin as a critical mediator of NF-κB signaling, which increases chromatin accessibility in the vicinity of NF-κB target genes in response to inflammation. Rad21 is required for normal differentiation, but limits self-renewal of hematopoietic stem cells (HSCs) during aging and inflammation in an NF-κB-dependent manner. HSCs from aged mice fail to down-regulate Rad21/cohesin and inflammation/differentiation signals in the resolution phase of inflammation. Inhibition of cohesin/NF-κB reverts hypersensitivity of aged HSPCs to inflammation-induced differentiation and myeloid-biased HSCs with disrupted/reduced expression of Rad21/cohesin are increasingly selected during aging. Together, Rad21/cohesin-mediated NF-κB signaling limits HSPC function during aging and selects for cohesin-deficient HSCs with myeloid-skewed differentiation.
Collapse
Affiliation(s)
- Zhiyang Chen
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Elias Moris Amro
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Friedrich Becker
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Martin Hölzer
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, Jena, Germany
- European Virus Bioinformatics Center (EVBC), Jena, Germany
| | | | | | - Bing Han
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Simone Di Sanzo
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Yulin Chen
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Duozhuang Tang
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Si Tao
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Ronny Haenold
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
- Matthias Schleiden Institute for Genetics, Bioinformatics and Molecular Botany, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Marco Groth
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Vasily S Romanov
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | | | - Johann M Kraus
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Manja Marz
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, Jena, Germany
- European Virus Bioinformatics Center (EVBC), Jena, Germany
| | - Alessandro Ori
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Francesco Neri
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Yohei Morita
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - K Lenhard Rudolph
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
- Faculty of Medicine, Friedrich-Schiller-University, Jena, Germany
| |
Collapse
|