1
|
Wang X, Guillem-Marti J, Kumar S, Lee DS, Cabrerizo-Aguado D, Werther R, Alamo KAE, Zhao YT, Nguyen A, Kopyeva I, Huang B, Li J, Hao Y, Li X, Brizuela-Velasco A, Murray A, Gerben S, Roy A, DeForest CA, Springer T, Ruohola-Baker H, Cooper JA, Campbell MG, Manero JM, Ginebra MP, Baker D. De Novo Design of Integrin α5β1 Modulating Proteins for Regenerative Medicine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600123. [PMID: 38979380 PMCID: PMC11230231 DOI: 10.1101/2024.06.21.600123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Integrin α5β1 is crucial for cell attachment and migration in development and tissue regeneration, and α5β1 binding proteins could have considerable utility in regenerative medicine and next-generation therapeutics. We use computational protein design to create de novo α5β1-specific modulating miniprotein binders, called NeoNectins, that bind to and stabilize the open state of α5β1. When immobilized onto titanium surfaces and throughout 3D hydrogels, the NeoNectins outperform native fibronectin and RGD peptide in enhancing cell attachment and spreading, and NeoNectin-grafted titanium implants outperformed fibronectin and RGD-grafted implants in animal models in promoting tissue integration and bone growth. NeoNectins should be broadly applicable for tissue engineering and biomedicine.
Collapse
Affiliation(s)
- Xinru Wang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jordi Guillem-Marti
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya - BarcelonaTech (UPC), Barcelona, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain
| | - Saurav Kumar
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - David S Lee
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Daniel Cabrerizo-Aguado
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya - BarcelonaTech (UPC), Barcelona, Spain
| | - Rachel Werther
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Yan Ting Zhao
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Adam Nguyen
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Graduate Program in Biological Physics, Structure and Design, University of Washington, Seattle, WA, USA
| | - Irina Kopyeva
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Buwei Huang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jing Li
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Yuxin Hao
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Xinting Li
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Aritza Brizuela-Velasco
- DENS-ia Research Group, Faculty of Health Sciences, Miguel de Cervantes European University, Valladolid, Spain
| | - Analisa Murray
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Stacey Gerben
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Anindya Roy
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Cole A DeForest
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
- Department of Chemistry, University of Washington, Seattle, WA, USA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA, USA
| | - Timothy Springer
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Hannele Ruohola-Baker
- Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jonathan A Cooper
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Melody G Campbell
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jose Maria Manero
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya - BarcelonaTech (UPC), Barcelona, Spain
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya - BarcelonaTech (UPC), Barcelona, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain
- Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
2
|
Chaudhari JK, Pant S, Jha R, Pathak RK, Singh DB. Biological big-data sources, problems of storage, computational issues, and applications: a comprehensive review. Knowl Inf Syst 2024; 66:3159-3209. [DOI: 10.1007/s10115-023-02049-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/12/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2025]
|
3
|
Xu Q, Li CH, Tang CH, Huang XL, Wu LF, Zhou X, Lei SF, Deng FY. PKM2 is a Novel Osteoporosis-Associated Protein in Chinese. Endocr Res 2024; 49:92-105. [PMID: 38288985 DOI: 10.1080/07435800.2024.2310818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/16/2023] [Indexed: 04/24/2024]
Abstract
Purpose:Osteoporosis is characterized by low bone mineral density (BMD) and high risk of osteoporotic fracture (OF). Peripheral blood monocytes (PBM) can differentiate into osteoclasts to resorb bone. This study was to identify PBM-expressed proteins significant for osteoporosis in Chinese Han elderly population (>65 years), and focused on two phenotypes of osteoporosis: low BMD and OF. METHODS Label-free quantitative proteomics was employed to profile PBM proteome and to identify differentially expressed proteins (DEPs) between OF (N=27) vs. non-fractured (NF, N=24) subjects and between low BMD (N=12) vs. high BMD (N=12) subjects in women. Western blotting (WB) was conducted to validate differential expression, and ELISA to evaluate translational value for secretory protein of interest. RESULTS We discovered 59 DEPs with fold change (FC)>1.3 (P<1×10-5), and validated the significant up-regulation of pyruvate kinase isozyme 2 (PKM2) with osteoporosis (P<0.001). PKM2 protein upregulation with OF was replicated with PBM in men (P=0.04). Plasma PKM2 protein level was significantly elevated with OF in an independent sample (N=100, FC=1.68, P=0.01). Pursuant functional assays showed that extracellular PKM2 protein supplement not only promoted monocyte trans-endothelial migration, growth, and osteoclast differentiation (marker gene expression), but also inhibited osteoblast growth, differentiation (ALP gene expression), and activity. CONCLUSION The above findings suggest that PKM2 protein is a novel osteoporosis-associated functional protein in Chinese Han elderly population. It may serve as a risk biomarker and drug target for osteoporosis.
Collapse
Affiliation(s)
- Qing Xu
- School of Public Health, Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University; Center for Genetic Epidemiology and Genomics, Suzhou, Jiangsu, P. R. China
| | - Chun-Hui Li
- School of Public Health, Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University; Center for Genetic Epidemiology and Genomics, Suzhou, Jiangsu, P. R. China
| | - Chang-Hua Tang
- Department of Orthopedics, Sihong Hospital, Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Suqian, Jiangsu, P. R. China
| | - Xiao-Li Huang
- Department of Orthopedics, Sihong Hospital, Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Suqian, Jiangsu, P. R. China
| | - Long-Fei Wu
- School of Public Health, Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University; Center for Genetic Epidemiology and Genomics, Suzhou, Jiangsu, P. R. China
| | - Xu Zhou
- School of Public Health, Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University; Center for Genetic Epidemiology and Genomics, Suzhou, Jiangsu, P. R. China
| | - Shu-Feng Lei
- School of Public Health, Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University; Center for Genetic Epidemiology and Genomics, Suzhou, Jiangsu, P. R. China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases; MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, P. R. China
- Department of Orthopedics, Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou, Jiangsu, P. R. China
| | - Fei-Yan Deng
- School of Public Health, Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University; Center for Genetic Epidemiology and Genomics, Suzhou, Jiangsu, P. R. China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases; MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, P. R. China
| |
Collapse
|
4
|
Muneer A, Wang L, Xie L, Zhang F, Wu B, Mei L, Lenarcic EM, Feng EH, Song J, Xiong Y, Yu X, Wang C, Jain K, Strahl BD, Cook JG, Wan YY, Moorman NJ, Song H, Jin J, Chen X. Non-canonical function of histone methyltransferase G9a in the translational regulation of chronic inflammation. Cell Chem Biol 2023; 30:1525-1541.e7. [PMID: 37858336 PMCID: PMC11095832 DOI: 10.1016/j.chembiol.2023.09.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 06/21/2023] [Accepted: 09/20/2023] [Indexed: 10/21/2023]
Abstract
We report a novel translation-regulatory function of G9a, a histone methyltransferase and well-understood transcriptional repressor, in promoting hyperinflammation and lymphopenia; two hallmarks of endotoxin tolerance (ET)-associated chronic inflammatory complications. Using multiple approaches, we demonstrate that G9a interacts with multiple translation regulators during ET, particularly the N6-methyladenosine (m6A) RNA methyltransferase METTL3, to co-upregulate expression of certain m6A-modified mRNAs that encode immune-checkpoint and anti-inflammatory proteins. Mechanistically, G9a promotes m6A methyltransferase activity of METTL3 at translational/post-translational level by regulating its expression, its methylation, and its cytosolic localization during ET. Additionally, from a broader view extended from the G9a-METTL3-m6A translation regulatory axis, our translatome proteomics approach identified numerous "G9a-translated" proteins that unite the networks associated with inflammation dysregulation, T cell dysfunction, and systemic cytokine response. In sum, we identified a previously unrecognized function of G9a in protein-specific translation that can be leveraged to treat ET-related chronic inflammatory diseases.
Collapse
Affiliation(s)
- Adil Muneer
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Li Wang
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ling Xie
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Feng Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bing Wu
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Liu Mei
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Erik M Lenarcic
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Emerald Hillary Feng
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yan Xiong
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xufen Yu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Charles Wang
- Center for Genomics, Division of Microbiology & Molecular Genetics, Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92350, USA
| | - Kanishk Jain
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian D Strahl
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jeanette Gowen Cook
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yisong Y Wan
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nathaniel John Moorman
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xian Chen
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
5
|
Negri VA, Louis B, Zijl S, Ganier C, Philippeos C, Ali S, Reynolds G, Haniffa M, Watt FM. Single-cell RNA sequencing of human epidermis identifies Lunatic fringe as a novel regulator of the stem cell compartment. Stem Cell Reports 2023; 18:2047-2055. [PMID: 37832539 PMCID: PMC10679657 DOI: 10.1016/j.stemcr.2023.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 10/15/2023] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) of human skin provides a tool for validating observations from in vitro experimental models. By analyzing a published dataset of healthy adult epidermis, we confirm that the basal epidermal layer is heterogeneous, and three subpopulations of non-dividing cells can be distinguished. We show that Delta-like ligand 1 (DLL1) is expressed in a subset of basal cells previously identified as stem cells in cultured human keratinocytes and map the distribution of other Notch ligands and receptors to specific epidermal cell compartments. Although DLL1 is expressed at low levels, it is expressed in the same cell state as the Notch regulator, Lunatic -fringe (LFNG, O-fucosylpeptide 3-beta-N-acetylglucosaminyltransferase). Overexpression of LFNG amplifies the effects of DLL1 in cultured keratinocytes, increasing proliferation and colony-forming ability. We conclude that using scRNA-seq resources from healthy human skin not only validates previous experimental data but allows formulation of testable new hypotheses.
Collapse
Affiliation(s)
- Victor Augusti Negri
- King's College London Centre for Stem Cells and Regenerative Medicine, Guy's Hospital, London, UK
| | - Blaise Louis
- King's College London Centre for Stem Cells and Regenerative Medicine, Guy's Hospital, London, UK
| | - Sebastiaan Zijl
- King's College London Centre for Stem Cells and Regenerative Medicine, Guy's Hospital, London, UK
| | - Clarisse Ganier
- King's College London Centre for Stem Cells and Regenerative Medicine, Guy's Hospital, London, UK
| | - Christina Philippeos
- King's College London Centre for Stem Cells and Regenerative Medicine, Guy's Hospital, London, UK
| | - Shahnawaz Ali
- King's College London Centre for Stem Cells and Regenerative Medicine, Guy's Hospital, London, UK
| | - Gary Reynolds
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Fiona M Watt
- King's College London Centre for Stem Cells and Regenerative Medicine, Guy's Hospital, London, UK; Directors' Research Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
6
|
Kumar S, Stainer A, Dubrulle J, Simpkins C, Cooper JA. Cas phosphorylation regulates focal adhesion assembly. eLife 2023; 12:e90234. [PMID: 37489578 PMCID: PMC10435235 DOI: 10.7554/elife.90234] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/19/2023] [Indexed: 07/26/2023] Open
Abstract
Integrin-mediated cell attachment rapidly induces tyrosine kinase signaling. Despite years of research, the role of this signaling in integrin activation and focal adhesion assembly is unclear. We provide evidence that the Src-family kinase (SFK) substrate Cas (Crk-associated substrate, p130Cas, BCAR1) is phosphorylated and associated with its Crk/CrkL effectors in clusters that are precursors of focal adhesions. The initial phospho-Cas clusters contain integrin β1 in its inactive, bent closed, conformation. Later, phospho-Cas and total Cas levels decrease as integrin β1 is activated and core focal adhesion proteins including vinculin, talin, kindlin, and paxillin are recruited. Cas is required for cell spreading and focal adhesion assembly in epithelial and fibroblast cells on collagen and fibronectin. Cas cluster formation requires Cas, Crk/CrkL, SFKs, and Rac1 but not vinculin. Rac1 provides positive feedback onto Cas through reactive oxygen, opposed by negative feedback from the ubiquitin proteasome system. The results suggest a two-step model for focal adhesion assembly in which clusters of phospho-Cas, effectors and inactive integrin β1 grow through positive feedback prior to integrin activation and recruitment of core focal adhesion proteins.
Collapse
Affiliation(s)
- Saurav Kumar
- Fred Hutchinson Cancer CenterSeattleUnited States
| | | | | | | | | |
Collapse
|
7
|
Negri VA, Watt FM. Understanding Human Epidermal Stem Cells at Single-Cell Resolution. J Invest Dermatol 2022; 142:2061-2067. [PMID: 35570025 PMCID: PMC9826868 DOI: 10.1016/j.jid.2022.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/20/2022] [Accepted: 04/01/2022] [Indexed: 01/13/2023]
Abstract
The human epidermis is one of the first tissues in which the existence of stem cells was recognized and is one of the few in which ex vivo expansion for tissue repair is established clinically. Nevertheless, the nature of stem cells has been elusive. Using clonal growth assays of cultured keratinocytes as a quantitative measure of their abundance, several candidate stem cell markers have been described. Recently, the volume and quality of single-cell RNA-sequencing datasets have increased exponentially, providing new opportunities to explore the nature of epidermal stem cells and test the validity of in vitro experimental models.
Collapse
Affiliation(s)
- Victor Augusti Negri
- Centre for Gene Therapy and Regenerative Medicine, Faculty of Life Sciences & Medicine, School of Basic & Medical Biosciences, King’s College London, London, United Kingdom
| | - Fiona M. Watt
- Centre for Gene Therapy and Regenerative Medicine, Faculty of Life Sciences & Medicine, School of Basic & Medical Biosciences, King’s College London, London, United Kingdom,Directors' Research Unit, European Molecular Biology Laboratory, Heidelberg, Germany,Correspondence: Fiona M. Watt, Centre for Gene Therapy and Regenerative Medicine, King’s College London, Floor 28, Tower Wing, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United Kingdom.
| |
Collapse
|
8
|
Kennedy J, Whiteaker JR, Ivey RG, Burian A, Chowdhury S, Tsai CF, Liu T, Lin C, Murillo OD, Lundeen RA, Jones LA, Gafken PR, Longton G, Rodland KD, Skates SJ, Landua J, Wang P, Lewis MT, Paulovich AG. Internal Standard Triggered-Parallel Reaction Monitoring Mass Spectrometry Enables Multiplexed Quantification of Candidate Biomarkers in Plasma. Anal Chem 2022; 94:9540-9547. [PMID: 35767427 PMCID: PMC9280723 DOI: 10.1021/acs.analchem.1c04382] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Despite advances in proteomic technologies, clinical translation of plasma biomarkers remains low, partly due to a major bottleneck between the discovery of candidate biomarkers and costly clinical validation studies. Due to a dearth of multiplexable assays, generally only a few candidate biomarkers are tested, and the validation success rate is accordingly low. Previously, mass spectrometry-based approaches have been used to fill this gap but feature poor quantitative performance and were generally limited to hundreds of proteins. Here, we demonstrate the capability of an internal standard triggered-parallel reaction monitoring (IS-PRM) assay to greatly expand the numbers of candidates that can be tested with improved quantitative performance. The assay couples immunodepletion and fractionation with IS-PRM and was developed and implemented in human plasma to quantify 5176 peptides representing 1314 breast cancer biomarker candidates. Characterization of the IS-PRM assay demonstrated the precision (median % CV of 7.7%), linearity (median R2 > 0.999 over 4 orders of magnitude), and sensitivity (median LLOQ < 1 fmol, approximately) to enable rank-ordering of candidate biomarkers for validation studies. Using three plasma pools from breast cancer patients and three control pools, 893 proteins were quantified, of which 162 candidate biomarkers were verified in at least one of the cancer pools and 22 were verified in all three cancer pools. The assay greatly expands capabilities for quantification of large numbers of proteins and is well suited for prioritization of viable candidate biomarkers.
Collapse
Affiliation(s)
- Jacob
J. Kennedy
- Clinical
Research Division, Fred Hutchinson Cancer
Research Center, Seattle, Washington 98109, United States
| | - Jeffrey R. Whiteaker
- Clinical
Research Division, Fred Hutchinson Cancer
Research Center, Seattle, Washington 98109, United States
| | - Richard G. Ivey
- Clinical
Research Division, Fred Hutchinson Cancer
Research Center, Seattle, Washington 98109, United States
| | - Aura Burian
- Clinical
Research Division, Fred Hutchinson Cancer
Research Center, Seattle, Washington 98109, United States
| | - Shrabanti Chowdhury
- Department
of Genetics and Genomic Sciences and Icahn Institute for Data Science
and Genomic Technology, Icahn School of
Medicine at Mount Sinai, New York, New York 10029, United States
| | - Chia-Feng Tsai
- Biological
Sciences Division, Pacific Northwest National
Laboratory, Richland, Washington 99352, United States
| | - Tao Liu
- Biological
Sciences Division, Pacific Northwest National
Laboratory, Richland, Washington 99352, United States
| | - ChenWei Lin
- Clinical
Research Division, Fred Hutchinson Cancer
Research Center, Seattle, Washington 98109, United States
| | - Oscar D. Murillo
- Clinical
Research Division, Fred Hutchinson Cancer
Research Center, Seattle, Washington 98109, United States
| | - Rachel A. Lundeen
- Clinical
Research Division, Fred Hutchinson Cancer
Research Center, Seattle, Washington 98109, United States
| | - Lisa A. Jones
- Proteomics
and Metabolomics Shared Resources, Fred
Hutchinson Cancer Research Center, Seattle, Washington 98109, United States
| | - Philip R. Gafken
- Proteomics
and Metabolomics Shared Resources, Fred
Hutchinson Cancer Research Center, Seattle, Washington 98109, United States
| | - Gary Longton
- Public
Health Sciences Division, Fred Hutchinson
Cancer Research Center, Seattle, Washington 98109, United States
| | - Karin D. Rodland
- Biological
Sciences Division, Pacific Northwest National
Laboratory, Richland, Washington 99352, United States
| | - Steven J. Skates
- MGH
Biostatistics Center, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - John Landua
- Lester
and Sue Smith Breast Center, Baylor College
of Medicine, Houston, Texas 77030, United States
| | - Pei Wang
- Department
of Genetics and Genomic Sciences, Mount
Sinai Hospital, New York, New York 10065, United States
| | - Michael T. Lewis
- Lester
and Sue Smith Breast Center, Baylor College
of Medicine, Houston, Texas 77030, United States
| | - Amanda G. Paulovich
- Clinical
Research Division, Fred Hutchinson Cancer
Research Center, Seattle, Washington 98109, United States,Phone: 206-667-1912. . Fax: 206-667-2277
| |
Collapse
|
9
|
Zijl S, Salameti V, Louis B, Negri VA, Watt FM. Dynamic regulation of human epidermal differentiation by adhesive and mechanical forces. Curr Top Dev Biol 2022; 150:129-148. [DOI: 10.1016/bs.ctdb.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
10
|
Heinzlmeir S, Müller S. Selectivity aspects of activity-based (chemical) probes. Drug Discov Today 2021; 27:519-528. [PMID: 34728376 DOI: 10.1016/j.drudis.2021.10.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 09/20/2021] [Accepted: 10/27/2021] [Indexed: 12/19/2022]
Abstract
Selective chemical modulators are ideal tools to study the function of a protein. Yet, the poor ligandability of many proteins has hampered the development of specific chemical probes for numerous protein classes. Tools, such as covalent inhibitors and activity-based protein profiling, have enhanced our understanding of thus-far difficult-to-target proteins and have enabled correct assessment of the selectivity of small-molecule modulators. This also requires deeper knowledge of compound and target site reactivity, evaluation of binding to noncovalent targets and protein turnover. The availability of highly selective chemical probes, the evolution of activity-based probes, and the development of profiling methods will open a new era of drugging the undruggable proteome.
Collapse
Affiliation(s)
- Stephanie Heinzlmeir
- Technical University of Munich, 85354 Freising, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Susanne Müller
- Structural Genomics Consortium, Goethe University Frankfurt, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Strabe 15, 60438 Frankfurt am Main, Germany; Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Strabe 9, 60438 Frankfurt, Germany; The Chemical Probes Portal, The Institute of Cancer Research, London SM2 5NG, UK.
| |
Collapse
|
11
|
Jaswal S, Anand V, Ali SA, Jena MK, Kumar S, Kaushik JK, Mohanty AK. TMT based deep proteome analysis of buffalo mammary epithelial cells and identification of novel protein signatures during lactogenic differentiation. FASEB J 2021; 35:e21621. [PMID: 33977573 DOI: 10.1096/fj.202002476rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 01/04/2023]
Abstract
The lactating mammary gland harbours numerous matured alveoli with their lumen surrounded by differentiated mammary epithelial cells (MECs), which are exclusively involved in milk synthesis and secretion. Buffalo (Bubalus bubalis) is the second major milk-producing animal, and its physiology is different from cattle. The complete protein machinery involved in MECs differentiation is still not defined in ruminants, in particular, buffalo. Therefore, we have studied the differential expression of regulated proteins in the in vitro grown buffalo MECs (BuMECs) at different time points (on 3, 6, 12, and 15 days) of their differentiation in the presence of lactogenic hormones. TMT-based MS analysis identified 4,934 proteins; of them, 681 were differentially expressed proteins (DEPs). The principal component analysis suggested a highly heterogeneous expression of DEPs at the four-time points of hormone treatment, with most of them (307) attained the highest expression on 12 days. Bioinformatics analysis revealed the association of DEPs with 24 KEGG pathways. We observed few new proteins, namely ABCA13, IVL, VPS37, CZIB, RFX7, Rab5, TTLL12, SMEK1, GDI2, and TMEM131 in BuMECs. The function of one of the highly upregulated proteins, namely involucrin in the differentiation of BuMECs was confirmed based on biochemical inhibition assay. The results further conclude that the proteins with higher abundance can be considered as the potential biomarkers for differentiation, and they may have a significant association with the lactation process in buffalo too. The proteome dataset obtained can be used to understand the species-specific variations among other lactating animals.
Collapse
Affiliation(s)
- Shalini Jaswal
- Proteomics and Cell Biology Lab, Animal Biotechnology Centre, National Dairy Research Institute (NDRI), Karnal, India
| | - Vijay Anand
- Department of Veterinary Physiology and Biochemistry, Veterinary College and Research Institute (TANUVAS), Orathanadu, India
| | - Syed Azmal Ali
- Proteomics and Cell Biology Lab, Animal Biotechnology Centre, National Dairy Research Institute (NDRI), Karnal, India
| | - Manoj K Jena
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India
| | - Sudarshan Kumar
- Proteomics and Cell Biology Lab, Animal Biotechnology Centre, National Dairy Research Institute (NDRI), Karnal, India
| | - Jai K Kaushik
- Proteomics and Cell Biology Lab, Animal Biotechnology Centre, National Dairy Research Institute (NDRI), Karnal, India
| | - Ashok K Mohanty
- Proteomics and Cell Biology Lab, Animal Biotechnology Centre, National Dairy Research Institute (NDRI), Karnal, India
| |
Collapse
|
12
|
Wang L, Muneer A, Xie L, Zhang F, Wu B, Mei L, Lenarcic EM, Feng EH, Song J, Xiong Y, Yu X, Wang C, Gheorghe C, Torralba K, Cook JG, Wan YY, Moorman NJ, Song H, Jin J, Chen X. Novel gene-specific translation mechanism of dysregulated, chronic inflammation reveals promising, multifaceted COVID-19 therapeutics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.11.14.382416. [PMID: 33236014 PMCID: PMC7685324 DOI: 10.1101/2020.11.14.382416] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hyperinflammation and lymphopenia provoked by SARS-CoV-2-activated macrophages contribute to the high mortality of Coronavirus Disease 2019 (COVID-19) patients. Thus, defining host pathways aberrantly activated in patient macrophages is critical for developing effective therapeutics. We discovered that G9a, a histone methyltransferase that is overexpressed in COVID-19 patients with high viral load, activates translation of specific genes that induce hyperinflammation and impairment of T cell function or lymphopenia. This noncanonical, pro-translation activity of G9a contrasts with its canonical epigenetic function. In endotoxin-tolerant (ET) macrophages that mimic conditions which render patients with pre-existing chronic inflammatory diseases vulnerable to severe symptoms, our chemoproteomic approach with a biotinylated inhibitor of G9a identified multiple G9a-associated translation regulatory pathways that were upregulated by SARS-CoV-2 infection. Further, quantitative translatome analysis of ET macrophages treated progressively with the G9a inhibitor profiled G9a-translated proteins that unite the networks associated with viral replication and the SARS-CoV-2-induced host response in severe patients. Accordingly, inhibition of G9a-associated pathways produced multifaceted, systematic effects, namely, restoration of T cell function, mitigation of hyperinflammation, and suppression of viral replication. Importantly, as a host-directed mechanism, this G9a-targeted, combined therapeutics is refractory to emerging antiviral-resistant mutants of SARS-CoV-2, or any virus, that hijacks host responses.
Collapse
|
13
|
Schneider K, Nelson GM, Watson JL, Morf J, Dalglish M, Luh LM, Weber A, Bertolotti A. Protein Stability Buffers the Cost of Translation Attenuation following eIF2α Phosphorylation. Cell Rep 2020; 32:108154. [PMID: 32937139 PMCID: PMC7495045 DOI: 10.1016/j.celrep.2020.108154] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/25/2020] [Accepted: 08/25/2020] [Indexed: 01/28/2023] Open
Abstract
Phosphorylation of the translation initiation factor eIF2α is a rapid and vital response to many forms of stress, including protein-misfolding stress in the endoplasmic reticulum (ER stress). It is believed to cause a general reduction in protein synthesis while enabling translation of few transcripts. Such a reduction of protein synthesis comes with the threat of depleting essential proteins, a risk thought to be mitigated by its transient nature. Here, we find that translation attenuation is not uniform, with cytosolic and mitochondrial ribosomal subunits being prominently downregulated. Translation attenuation of these targets persists after translation recovery. Surprisingly, this occurs without a measurable decrease in ribosomal proteins. Explaining this conundrum, translation attenuation preferentially targets long-lived proteins, a finding not only demonstrated by ribosomal proteins but also observed at a global level. This shows that protein stability buffers the cost of translational attenuation, establishing an evolutionary principle of cellular robustness.
Collapse
Affiliation(s)
- Kim Schneider
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, United Kingdom.
| | - Geoffrey Michael Nelson
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, United Kingdom
| | - Joseph Luke Watson
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, United Kingdom
| | - Jörg Morf
- Wellcome - MRC Cambridge Stem Cell Institute, Puddicombe Way, Cambridge CB2 0AW, United Kingdom
| | - Maximillian Dalglish
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, United Kingdom
| | - Laura Martina Luh
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, United Kingdom
| | - Annika Weber
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, United Kingdom
| | - Anne Bertolotti
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, United Kingdom.
| |
Collapse
|
14
|
H2A Histone Family Member X (H2AX) Is Upregulated in Ovarian Cancer and Demonstrates Utility as a Prognostic Biomarker in Terms of Overall Survival. J Clin Med 2020; 9:jcm9092844. [PMID: 32887437 PMCID: PMC7565050 DOI: 10.3390/jcm9092844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/25/2020] [Accepted: 08/30/2020] [Indexed: 12/12/2022] Open
Abstract
Background: H2AX can be of prognostic value in breast cancer, since in advanced stage patients with high levels, there was an association with worse overall survival (OS). However, the clinical relevance of H2AX in ovarian cancer (OC) remains to be elucidated. Methods: OC H2AX expression studied using the TCGA/GTEX datasets. Subsequently, patients were classified as either high or low in terms of H2AX expression to compare OS and perform gene set enrichment. qRT-PCR validated in-silico H2AX findings followed by immunohistochemistry on a tissue microarray. The association between single nucleotide polymorphisms in the area of H2AX; prevalence and five-year OC survival was tested in samples from the UK Biobank. Results: H2AX was significantly overexpressed in OCs compared to normal tissues, with higher expression associated with better OS (p = 0.010). Gene Set Enrichment Analysis demonstrated gene sets involved in G2/M checkpoint, DNA repair mTORC1 signalling were enriched in the H2AX highly expressing OCs. Polymorphisms in the area around the gene were associated with both OC prevalence (rs72997349-C, p = 0.005) and worse OS (rs10790282-G, p = 0.011). Finally, we demonstrated that H2AX gene expression correlated with γ-H2AX staining in vitro. Conclusions: Our findings suggest that H2AX can be a novel prognostic biomarker for OC.
Collapse
|
15
|
Runau F, Arshad A, Isherwood JD, Sandhu JK, Ng LL, Dennison AR, Jones DJL. Proteomic Characterization of Circulating Molecular Perturbations Associated With Pancreatic Adenocarcinoma Following Intravenous ω-3 Fatty Acid and Gemcitabine Administration: A Pilot Study. JPEN J Parenter Enteral Nutr 2020; 45:738-750. [PMID: 32716569 DOI: 10.1002/jpen.1952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND Administration of intravenous ω-3 fatty acid (ω-3FA) in advanced pancreatic adenocarcinoma patients receiving gemcitabine chemotherapy shows disease stabilization and improved progression-free survival. Using high-definition plasma proteomics, the underlying biological mechanisms responsible for these clinical effects are investigated. METHODS AND RESULTS A pilot study involving plasma that was collected at baseline from 13 patients with histologically confirmed, unresectable pancreatic adenocarcinoma (baseline group) after 1-month treatment with intravenous gemcitabine and ω-3FA (treatment group) and intravenous gemcitabine only (control group) and was prepared for proteomic analysis. A 2-arm study comparing baseline vs treatment and treatment vs control was performed. Proteins were isolated from plasma with extensive immunodepletion, then digested and labeled with isobaric tandem mass tag peptide tags. Samples were then combined, fractionated, and injected into a QExactive-Orbitrap Mass-Spectrometer and analyzed on Proteome Discoverer and Scaffold with ensuing bioinformatics analysis. Selective reaction monitoring analysis was performed for verification. In total, 3476 proteins were identified. Anti-inflammatory markers (C-reactive protein, haptoglobin, and serum amyloid-A1) were reduced in the treatment group. Enrichment analysis showed angiogenesis downregulation, complement immune systems upregulation, and epigenetic modifications on histones. Pathway analysis identified direct action via the Pi3K-AKT pathway. Serum amyloid-A1 significantly reduced (P < .001) as a potential biomarker of efficacy for ω-3FA. CONCLUSIONS This pilot study demonstrates administration of ω-3FA has potential anti-inflammatory, antiangiogenic, and proapoptotic effects via direct interaction with cancer-signaling pathways in patients with advanced pancreatic adenocarcinoma. Further studies in a larger sample size is required to validate the clinical correlation found in this preliminary study.
Collapse
Affiliation(s)
- Franscois Runau
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK.,Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Ali Arshad
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - John D Isherwood
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Jatinderpal K Sandhu
- Department of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - Leong L Ng
- Department of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - Ashley R Dennison
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Donald J L Jones
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK.,Department of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| |
Collapse
|
16
|
Tinti M, Güther MLS, Crozier TWM, Lamond AI, Ferguson MAJ. Proteome turnover in the bloodstream and procyclic forms of Trypanosoma brucei measured by quantitative proteomics. Wellcome Open Res 2019; 4:152. [PMID: 31681858 PMCID: PMC6816455 DOI: 10.12688/wellcomeopenres.15421.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Cellular proteins vary significantly in both abundance and turnover rates. These parameters depend upon their rates of synthesis and degradation and it is useful to have access to data on protein turnover rates when, for example, designing genetic knock-down experiments or assessing the potential usefulness of covalent enzyme inhibitors. Little is known about the nature and regulation of protein turnover in Trypanosoma brucei, the etiological agent of human and animal African trypanosomiasis. Methods: To establish baseline data on T. brucei proteome turnover, a Stable Isotope Labelling with Amino acids in Cell culture (SILAC)-based mass spectrometry analysis was performed to reveal the synthesis and degradation profiles for thousands of proteins in the bloodstream and procyclic forms of this parasite. Results: This analysis revealed a slower average turnover rate of the procyclic form proteome relative to the bloodstream proteome. As expected, many of the proteins with the fastest turnover rates have functions in the cell cycle and in the regulation of cytokinesis in both bloodstream and procyclic forms. Moreover, the cellular localization of T. brucei proteins correlates with their turnover, with mitochondrial and glycosomal proteins exhibiting slower than average turnover rates. Conclusions: The intention of this study is to provide the trypanosome research community with a resource for protein turnover data for any protein or group of proteins. To this end, bioinformatic analyses of these data are made available via an open-access web resource with data visualization functions.
Collapse
Affiliation(s)
- Michele Tinti
- The Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, UK
| | - Maria Lucia S Güther
- The Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, UK
| | - Thomas W M Crozier
- The Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, UK.,Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK.,Department of Medicine, Cambridge Institute for Medical Research, Cambridge, UK
| | - Angus I Lamond
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK
| | - Michael A J Ferguson
- The Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, UK
| |
Collapse
|
17
|
Bensaddek D, Nicolas A, Lamond AI. Signal enhanced proteomics: a biological perspective on dissecting the functional organisation of cell proteomes. Curr Opin Chem Biol 2018; 48:114-122. [PMID: 30551035 DOI: 10.1016/j.cbpa.2018.10.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/07/2018] [Accepted: 10/10/2018] [Indexed: 01/28/2023]
Abstract
Proteomes are highly dynamic and can respond rapidly to environmental and cellular signals. Within cells, proteins often form distinct pools with different functions and properties. However, in quantitative proteomics studies it is common to measure averaged values for proteins that do not reflect variations that may occur between different protein isoforms, different subcellular compartments, or in cells at different cell cycle stages and so on. Here we review experimental approaches that can be used to enhance the signal from specific pools of protein that may otherwise be obscured through averaging across protein populations. This signal enhancement can help to reveal functions associated with specific protein pools, providing insight into the regulation of cellular processes. We review different strategies for proteomic signal enhancement, with a focus on the analysis of protein pools in different subcellular locations. We describe how MS-based proteome analyses can be combined with a general physico-chemical cell fractionation procedure that can be applied to many cultured cell lines.
Collapse
Affiliation(s)
- Dalila Bensaddek
- Laboratory of Quantitative Proteomics, Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Armel Nicolas
- Laboratory of Quantitative Proteomics, Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Angus I Lamond
- Laboratory of Quantitative Proteomics, Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| |
Collapse
|