1
|
Vora S, Andrew A, Kumar RP, Nazareth D, Bonfim-Melo A, Lim Y, Ong XY, Fernando M, He Y, Hooper JD, McMillan NA, Urosevic J, Travers J, Saeh J, Kumar S, Jones MJ, Gabrielli B. Aurora B inhibitors promote RB hypophosphorylation and senescence independent of p53-dependent CDK2/4 inhibition. Cell Death Dis 2024; 15:810. [PMID: 39521795 PMCID: PMC11550316 DOI: 10.1038/s41419-024-07204-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/27/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Aurora B kinase (AURKB) inhibitors have been trialled in a range of different tumour types but are not approved for any indication. Expression of the human papilloma virus (HPV) oncogenes and loss of retinoblastoma (RB) protein function has been reported to increase sensitivity to AURKB inhibitors but the mechanism of their contribution to sensitivity is poorly understood. Two commonly reported outcomes of AURKB inhibition are polyploidy and senescence, although their relationship is unclear. Here we have investigated the major cellular targets of the HPV E6 and E7, p53 and RB, to determine their contribution to AURKB inhibitor induced polyploidy and senescence. We demonstrate that polyploidy is a universal feature of AURKB inhibitor treatment in all cell types including normal primary cells, but the subsequent outcomes are controlled by RB and p53. We demonstrate that p53 by regulating p21 expression is required for an initial cell cycle arrest by inhibiting both CDK2 and CDK4 activity, but this arrest is only triggered after cells have undergone two failed mitosis and cytokinesis. However, cells can enter senescence in the absence of p53. RB is essential for AURKB inhibitor-induced senescence. AURKB inhibitor induces rapid hypophosphorylation of RB independent of inhibition of CDK2 or CDK4 kinases and p53. This work demonstrates that p53 activation determines the timing of senescence onset, but RB is indispensable for senescence.
Collapse
Affiliation(s)
- Shivam Vora
- Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Ariel Andrew
- Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | | | - Deborah Nazareth
- Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Alexis Bonfim-Melo
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Yoon Lim
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - Xin Yee Ong
- Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Madushan Fernando
- Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Yaowu He
- Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - John D Hooper
- Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Nigel Aj McMillan
- Menzies Health Institute Queensland and School of Medical Science, Griffith University, Gold Coast, QLD, Australia
| | - Jelena Urosevic
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Jon Travers
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Jamal Saeh
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - Mathew Jk Jones
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Brian Gabrielli
- Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
2
|
Yun S, Noh M, Yu J, Kim HJ, Hui CC, Lee H, Son JE. Unlocking biological mechanisms with integrative functional genomics approaches. Mol Cells 2024; 47:100092. [PMID: 39019219 PMCID: PMC11345568 DOI: 10.1016/j.mocell.2024.100092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024] Open
Abstract
Reverse genetics offers precise functional insights into genes through the targeted manipulation of gene expression followed by phenotypic assessment. While these approaches have proven effective in model organisms such as Saccharomyces cerevisiae, large-scale genetic manipulations in human cells were historically unfeasible due to methodological limitations. However, recent advancements in functional genomics, particularly clustered regularly interspaced short palindromic repeats (CRISPR)-based screening technologies and next-generation sequencing platforms, have enabled pooled screening technologies that allow massively parallel, unbiased assessments of biological phenomena in human cells. This review provides a comprehensive overview of cutting-edge functional genomic screening technologies applicable to human cells, ranging from short hairpin RNA screens to modern CRISPR screens. Additionally, we explore the integration of CRISPR platforms with single-cell approaches to monitor gene expression, chromatin accessibility, epigenetic regulation, and chromatin architecture following genetic perturbations at the omics level. By offering an in-depth understanding of these genomic screening methods, this review aims to provide insights into more targeted and effective strategies for genomic research and personalized medicine.
Collapse
Affiliation(s)
- Sehee Yun
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| | - Minsoo Noh
- Department of Life Sciences, Korea University, Seoul 02841, Korea; Department of Internal Medicine and Laboratory of Genomics and Translational Medicine, Gachon University College of Medicine, Incheon 21565, Korea
| | - Jivin Yu
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| | - Hyeon-Jai Kim
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| | - Chi-Chung Hui
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Hunsang Lee
- Department of Life Sciences, Korea University, Seoul 02841, Korea.
| | - Joe Eun Son
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Korea.
| |
Collapse
|
3
|
Jing Y, Jiang X, Ji Q, Wu Z, Wang W, Liu Z, Guillen-Garcia P, Esteban CR, Reddy P, Horvath S, Li J, Geng L, Hu Q, Wang S, Belmonte JCI, Ren J, Zhang W, Qu J, Liu GH. Genome-wide CRISPR activation screening in senescent cells reveals SOX5 as a driver and therapeutic target of rejuvenation. Cell Stem Cell 2023; 30:1452-1471.e10. [PMID: 37832549 DOI: 10.1016/j.stem.2023.09.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 08/04/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023]
Abstract
Our understanding of the molecular basis for cellular senescence remains incomplete, limiting the development of strategies to ameliorate age-related pathologies by preventing stem cell senescence. Here, we performed a genome-wide CRISPR activation (CRISPRa) screening using a human mesenchymal precursor cell (hMPC) model of the progeroid syndrome. We evaluated targets whose activation antagonizes cellular senescence, among which SOX5 outperformed as a top hit. Through decoding the epigenomic landscapes remodeled by overexpressing SOX5, we uncovered its role in resetting the transcription network for geroprotective genes, including HMGB2. Mechanistically, SOX5 binding elevated the enhancer activity of HMGB2 with increased levels of H3K27ac and H3K4me1, raising HMGB2 expression so as to promote rejuvenation. Furthermore, gene therapy with lentiviruses carrying SOX5 or HMGB2 rejuvenated cartilage and alleviated osteoarthritis in aged mice. Our study generated a comprehensive list of rejuvenators, pinpointing SOX5 as a potent driver for rejuvenation both in vitro and in vivo.
Collapse
Affiliation(s)
- Yaobin Jing
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaoyu Jiang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Qianzhao Ji
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Zeming Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Wei Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Zunpeng Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Pedro Guillen-Garcia
- Department of Traumatology and Research Unit, Clinica CEMTRO, 28035 Madrid, Spain
| | - Concepcion Rodriguez Esteban
- Altos Labs, Inc., San Diego, CA 94022, USA; Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Pradeep Reddy
- Altos Labs, Inc., San Diego, CA 94022, USA; Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Steve Horvath
- Altos Labs, Inc., San Diego, CA 94022, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 10833, USA
| | - Jingyi Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Lingling Geng
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Qinchao Hu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510060, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510060, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Chongqing Renji Hospital, University of Chinese Academy of Sciences, Chongqing 400062, China
| | - Juan Carlos Izpisua Belmonte
- Altos Labs, Inc., San Diego, CA 94022, USA; Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jie Ren
- Key Laboratory of RNA Science and Engineering, CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100190, China.
| | - Weiqi Zhang
- Key Laboratory of RNA Science and Engineering, CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100190, China.
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100190, China; Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
4
|
Fernando M, Duijf PHG, Proctor M, Stevenson AJ, Ehmann A, Vora S, Skalamera D, Adams M, Gabrielli B. Dysregulated G2 phase checkpoint recovery pathway reduces DNA repair efficiency and increases chromosomal instability in a wide range of tumours. Oncogenesis 2021; 10:41. [PMID: 33993200 PMCID: PMC8124070 DOI: 10.1038/s41389-021-00329-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 04/06/2021] [Accepted: 04/20/2021] [Indexed: 11/12/2022] Open
Abstract
Defective DNA repair is being demonstrated to be a useful target in cancer treatment. Currently, defective repair is identified by specific gene mutations, however defective repair is a common feature of cancers without these mutations. DNA damage triggers cell cycle checkpoints that are responsible for co-ordinating cell cycle arrest and DNA repair. Defects in checkpoint signalling components such as ataxia telangiectasia mutated (ATM) occur in a low proportion of cancers and are responsible for reduced DNA repair and increased genomic instability. Here we have investigated the AURKA-PLK1 cell cycle checkpoint recovery pathway that is responsible for exit from the G2 phase cell cycle checkpoint arrest. We demonstrate that dysregulation of PP6 and AURKA maintained elevated PLK1 activation to promote premature exit from only ATM, and not ATR-dependent checkpoint arrest. Surprisingly, depletion of the B55α subunit of PP2A that negatively regulates PLK1 was capable of overcoming ATM and ATR checkpoint arrests. Dysregulation of the checkpoint recovery pathway reduced S/G2 phase DNA repair efficiency and increased genomic instability. We found a strong correlation between dysregulation of the PP6-AURKA-PLK1-B55α checkpoint recovery pathway with signatures of defective homologous recombination and increased chromosomal instability in several cancer types. This work has identified an unrealised source of G2 phase DNA repair defects and chromosomal instability that are likely to be sensitive to treatments targeting defective repair.
Collapse
Affiliation(s)
- Madushan Fernando
- Mater Research Institute-The University of Queensland, Brisbane, QLD, Australia
| | - Pascal H G Duijf
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Faculty of Health, School of Biomedical Sciences, Brisbane, QLD, Australia
- Centre for Data Science, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Martina Proctor
- Mater Research Institute-The University of Queensland, Brisbane, QLD, Australia
| | | | - Anna Ehmann
- Mater Research Institute-The University of Queensland, Brisbane, QLD, Australia
| | - Shivam Vora
- Mater Research Institute-The University of Queensland, Brisbane, QLD, Australia
| | - Dubravka Skalamera
- Mater Research Institute-The University of Queensland, Brisbane, QLD, Australia
| | - Mark Adams
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Faculty of Health, School of Biomedical Sciences, Brisbane, QLD, Australia
| | - Brian Gabrielli
- Mater Research Institute-The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
5
|
Rae J, Ferguson C, Ariotti N, Webb RI, Cheng HH, Mead JL, Riches JD, Hunter DJ, Martel N, Baltos J, Christopoulos A, Bryce NS, Cagigas ML, Fonseka S, Sayre ME, Hardeman EC, Gunning PW, Gambin Y, Hall TE, Parton RG. A robust method for particulate detection of a genetic tag for 3D electron microscopy. eLife 2021; 10:64630. [PMID: 33904409 PMCID: PMC8104959 DOI: 10.7554/elife.64630] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Genetic tags allow rapid localization of tagged proteins in cells and tissues. APEX, an ascorbate peroxidase, has proven to be one of the most versatile and robust genetic tags for ultrastructural localization by electron microscopy (EM). Here, we describe a simple method, APEX-Gold, which converts the diffuse oxidized diaminobenzidine reaction product of APEX into a silver/gold particle akin to that used for immunogold labelling. The method increases the signal-to-noise ratio for EM detection, providing unambiguous detection of the tagged protein, and creates a readily quantifiable particulate signal. We demonstrate the wide applicability of this method for detection of membrane proteins, cytoplasmic proteins, and cytoskeletal proteins. The method can be combined with different EM techniques including fast freezing and freeze substitution, focussed ion beam scanning EM, and electron tomography. Quantitation of expressed APEX-fusion proteins is achievable using membrane vesicles generated by a cell-free expression system. These membrane vesicles possess a defined quantum of signal, which can act as an internal standard for determination of the absolute density of expressed APEX-fusion proteins. Detection of fusion proteins expressed at low levels in cells from CRISPR-edited mice demonstrates the high sensitivity of the APEX-Gold method.
Collapse
Affiliation(s)
- James Rae
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Charles Ferguson
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Nicholas Ariotti
- Mark Wainwright Analytical Centre, University of New South Wales, Sydney, Australia.,School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Richard I Webb
- The University of Queensland, Centre for Microscopy and Microanalysis, Queensland, Australia
| | - Han-Hao Cheng
- The University of Queensland, Centre for Microscopy and Microanalysis, Queensland, Australia
| | - James L Mead
- The University of Queensland, Centre for Microscopy and Microanalysis, Queensland, Australia.,Division Microrobotics and Control Engineering, Department of Computing Science, University of Oldenburg, Oldenburg, Germany
| | - James D Riches
- Queensland University of Technology, Queensland, Australia
| | - Dominic Jb Hunter
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia.,EMBL Australia Node for Single Molecule Sciences, University of New South Wales, Sydney, Australia
| | - Nick Martel
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Joanne Baltos
- Monash Institute of Pharmaceutical Sciences, Monash University, Victoria, Australia
| | - Arthur Christopoulos
- Monash Institute of Pharmaceutical Sciences, Monash University, Victoria, Australia
| | - Nicole S Bryce
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | | | - Sachini Fonseka
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Marcel E Sayre
- The University of Queensland, Centre for Microscopy and Microanalysis, Queensland, Australia
| | - Edna C Hardeman
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Peter W Gunning
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Yann Gambin
- EMBL Australia Node for Single Molecule Sciences, University of New South Wales, Sydney, Australia
| | - Thomas E Hall
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Robert G Parton
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia.,The University of Queensland, Centre for Microscopy and Microanalysis, Queensland, Australia
| |
Collapse
|
6
|
Khan AH, Lin A, Wang RT, Bloom JS, Lange K, Smith DJ. Pooled analysis of radiation hybrids identifies loci for growth and drug action in mammalian cells. Genome Res 2020; 30:1458-1467. [PMID: 32878976 PMCID: PMC7605260 DOI: 10.1101/gr.262204.120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022]
Abstract
Genetic screens in mammalian cells commonly focus on loss-of-function approaches. To evaluate the phenotypic consequences of extra gene copies, we used bulk segregant analysis (BSA) of radiation hybrid (RH) cells. We constructed six pools of RH cells, each consisting of ∼2500 independent clones, and placed the pools under selection in media with or without paclitaxel. Low pass sequencing identified 859 growth loci, 38 paclitaxel loci, 62 interaction loci, and three loci for mitochondrial abundance at genome-wide significance. Resolution was measured as ∼30 kb, close to single-gene. Divergent properties were displayed by the RH-BSA growth genes compared to those from loss-of-function screens, refuting the balance hypothesis. In addition, enhanced retention of human centromeres in the RH pools suggests a new approach to functional dissection of these chromosomal elements. Pooled analysis of RH cells showed high power and resolution and should be a useful addition to the mammalian genetic toolkit.
Collapse
Affiliation(s)
- Arshad H Khan
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California 90095-1735, USA
| | - Andy Lin
- Office of Information Technology, UCLA, Los Angeles, California 90095-1557, USA
| | - Richard T Wang
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, California 90095-7088, USA
| | - Joshua S Bloom
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, California 90095-7088, USA
- Howard Hughes Medical Institute, David Geffen School of Medicine, UCLA, Los Angeles, California 90095-7088, USA
| | - Kenneth Lange
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, California 90095-7088, USA
| | - Desmond J Smith
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California 90095-1735, USA
| |
Collapse
|
7
|
Kumar A, Mali P. Mapping regulators of cell fate determination: Approaches and challenges. APL Bioeng 2020; 4:031501. [PMID: 32637855 PMCID: PMC7332300 DOI: 10.1063/5.0004611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/01/2020] [Indexed: 12/25/2022] Open
Abstract
Given the limited regenerative capacities of most organs, strategies are needed to efficiently generate large numbers of parenchymal cells capable of integration into the diseased organ. Although it was initially thought that terminally differentiated cells lacked the ability to transdifferentiate, it has since been shown that cellular reprogramming of stromal cells to parenchymal cells through direct lineage conversion holds great potential for the replacement of post-mitotic parenchymal cells lost to disease. To this end, an assortment of genetic, chemical, and mechanical cues have been identified to reprogram cells to different lineages both in vitro and in vivo. However, some key challenges persist that limit broader applications of reprogramming technologies. These include: (1) low reprogramming efficiencies; (2) incomplete functional maturation of derived cells; and (3) difficulty in determining the typically multi-factor combinatorial recipes required for successful transdifferentiation. To improve efficiency by comprehensively identifying factors that regulate cell fate, large scale genetic and chemical screening methods have thus been utilized. Here, we provide an overview of the underlying concept of cell reprogramming as well as the rationale, considerations, and limitations of high throughput screening methods. We next follow with a summary of unique hits that have been identified by high throughput screens to induce reprogramming to various parenchymal lineages. Finally, we discuss future directions of applying this technology toward human disease biology via disease modeling, drug screening, and regenerative medicine.
Collapse
Affiliation(s)
- Aditya Kumar
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
8
|
Varasteh Moradi S, Gagoski D, Mureev S, Walden P, McMahon KA, Parton RG, Johnston WA, Alexandrov K. Mapping Interactions among Cell-Free Expressed Zika Virus Proteins. J Proteome Res 2020; 19:1522-1532. [DOI: 10.1021/acs.jproteome.9b00771] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Shayli Varasteh Moradi
- CSIRO-QUT Synthetic Biology Alliance, Centre for Tropical Crops and Bio-commodities, Queensland University of Technology, Brisbane QLD 4001, Australia
| | - Dejan Gagoski
- Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane QLD 4072, Australia
| | - Sergey Mureev
- CSIRO-QUT Synthetic Biology Alliance, Centre for Tropical Crops and Bio-commodities, Queensland University of Technology, Brisbane QLD 4001, Australia
| | - Patricia Walden
- CSIRO-QUT Synthetic Biology Alliance, Centre for Tropical Crops and Bio-commodities, Queensland University of Technology, Brisbane QLD 4001, Australia
| | - Kerrie-Ann McMahon
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane QLD 4072, Australia
| | - Robert G. Parton
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane QLD 4072, Australia
- Centre for Microscopy and Microanalysis, The University of Queensland, St Lucia, Brisbane QLD 4072, Australia
| | - Wayne A. Johnston
- CSIRO-QUT Synthetic Biology Alliance, Centre for Tropical Crops and Bio-commodities, Queensland University of Technology, Brisbane QLD 4001, Australia
| | - Kirill Alexandrov
- CSIRO-QUT Synthetic Biology Alliance, Centre for Tropical Crops and Bio-commodities, Queensland University of Technology, Brisbane QLD 4001, Australia
| |
Collapse
|
9
|
Kabadi A, McDonnell E, Frank CL, Drowley L. Applications of Functional Genomics for Drug Discovery. SLAS DISCOVERY 2020; 25:823-842. [PMID: 32026742 DOI: 10.1177/2472555220902092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Many diseases, such as diabetes, autoimmune diseases, cancer, and neurological disorders, are caused by a dysregulation of a complex interplay of genes. Genome-wide association studies have identified thousands of disease-linked polymorphisms in the human population. However, detailing the causative gene expression or functional changes underlying those associations has been elusive in many cases. Functional genomics is an emerging field of research that aims to deconvolute the link between genotype and phenotype by making use of large -omic data sets and next-generation gene and epigenome editing tools to perturb genes of interest. Here we review how functional genomic tools can be used to better understand the biological interplay between genes, improve disease modeling, and identify novel drug targets. Incorporation of functional genomic capabilities into conventional drug development pipelines is predicted to expedite the development of first-in-class therapeutics.
Collapse
Affiliation(s)
- Ami Kabadi
- Element Genomics, a UCB company, Durham, NC, USA
| | | | | | | |
Collapse
|
10
|
Škalamera D, Stevenson AJ, Ehmann A, Ainger SA, Lanagan C, Sturm RA, Gabrielli B. Melanoma mutations modify melanocyte dynamics in co-culture with keratinocytes or fibroblasts. J Cell Sci 2019; 132:jcs.234716. [PMID: 31767623 DOI: 10.1242/jcs.234716] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 11/21/2019] [Indexed: 12/25/2022] Open
Abstract
Melanocytic cell interactions are integral to skin homeostasis, and affect the outcome of multiple diseases, including cutaneous pigmentation disorders and melanoma. By using automated-microscopy and machine-learning-assisted morphology analysis of primary human melanocytes in co-culture, we performed combinatorial interrogation of melanocyte genotypic variants and functional assessment of lentivirus-introduced mutations. Keratinocyte-induced melanocyte dendricity, an indicator of melanocyte differentiation, was reduced in the melanocortin 1 receptor (MC1R) R/R variant strain and by NRAS.Q61K and BRAF.V600E expression, while expression of CDK4.R24C and RAC1.P29S had no detectable effect. Time-lapse tracking of melanocytes in co-culture revealed dynamic interaction phenotypes and hyper-motile cell states that indicated that, in addition to the known role in activating mitogenic signalling, MEK-pathway-activating mutations may also allow melanocytes to escape keratinocyte control and increase their invasive potential. Expanding this combinatorial platform will identify other therapeutic target mutations and melanocyte genetic variants, as well as increase understanding of skin cell interactions.
Collapse
Affiliation(s)
- Dubravka Škalamera
- Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, 4102 QLD, Australia
| | - Alexander J Stevenson
- Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, 4102 QLD, Australia
| | - Anna Ehmann
- Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, 4102 QLD, Australia
| | - Stephen A Ainger
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, 4102 QLD, Australia
| | - Catherine Lanagan
- Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, 4102 QLD, Australia
| | - Richard A Sturm
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, 4102 QLD, Australia
| | - Brian Gabrielli
- Mater Research Institute, University of Queensland, Translational Research Institute, Brisbane, 4102 QLD, Australia
| |
Collapse
|
11
|
Ortega C, Abreu C, Oppezzo P, Correa A. Overview of High-Throughput Cloning Methods for the Post-genomic Era. Methods Mol Biol 2019; 2025:3-32. [PMID: 31267446 DOI: 10.1007/978-1-4939-9624-7_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The advent of new DNA sequencing technologies leads to a dramatic increase in the number of available genome sequences and therefore of target genes with potential for functional analysis. The insertion of these sequences into proper expression vectors requires a simple an efficient cloning method. In addition, when expressing a target protein, quite often it is necessary to evaluate different DNA constructs to achieve a soluble and homogeneous expression of the target with satisfactory yields. The development of new molecular methods made possible the cloning of a huge number of DNA sequences in a high-throughput manner, necessary for meeting the increasing demands for soluble protein expression and characterization. In this chapter several molecular methods suitable for high-throughput cloning are reviewed.
Collapse
Affiliation(s)
- Claudia Ortega
- Recombinant Protein Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Research Laboratory on Chronic Lymphocytic Leukemia, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Cecilia Abreu
- Recombinant Protein Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Molecular, Cellular and Animal Technology Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Pablo Oppezzo
- Recombinant Protein Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Research Laboratory on Chronic Lymphocytic Leukemia, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Agustín Correa
- Recombinant Protein Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay.
- Research Laboratory on Chronic Lymphocytic Leukemia, Institut Pasteur de Montevideo, Montevideo, Uruguay.
| |
Collapse
|
12
|
Hunter DJB, Bhumkar A, Giles N, Sierecki E, Gambin Y. Unexpected instabilities explain batch-to-batch variability in cell-free protein expression systems. Biotechnol Bioeng 2018; 115:1904-1914. [PMID: 29603735 DOI: 10.1002/bit.26604] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 02/22/2018] [Accepted: 03/26/2018] [Indexed: 01/09/2023]
Abstract
Cell-free methods of protein synthesis offer rapid access to expressed proteins. Though the amounts produced are generally only at a small scale, these are sufficient to perform protein-protein interaction assays and tests of enzymatic activity. As such they are valuable tools for the biochemistry and bioengineering community. However the most complex, eukaryotic cell-free systems are difficult to manufacture in house and can be prohibitively expensive to obtain from commercial sources. The Leishmania tarentolae system offers a relatively cheap alternative which is capable of producing difficult to express proteins, but which is simpler to produce in large scale. However, this system suffers from batch-to-batch variability, which has been accepted as a consequence of the complexity of the extracts. Here we show an unexpected origin for the variability observed and demonstrate that small variations in a single parameter can dramatically affect expression, such that minor pipetting errors can have major effects on yields. L. tarentolae cell-free lysate activity is shown to be more stable to changes in Mg2+ concentration at a lower ratio of feed solution to lysate in the reaction than typically used, and a higher Mg2+ optimum. These changes essentially eliminate batch-to-batch variability of L. tarentolae lysate activity and permit their full potential to be realized.
Collapse
Affiliation(s)
- Dominic J B Hunter
- EMBL Australia Node for Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia.,Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Akshay Bhumkar
- EMBL Australia Node for Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Nichole Giles
- EMBL Australia Node for Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Emma Sierecki
- EMBL Australia Node for Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Yann Gambin
- EMBL Australia Node for Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
13
|
Structural basis of TIR-domain-assembly formation in MAL- and MyD88-dependent TLR4 signaling. Nat Struct Mol Biol 2017; 24:743-751. [PMID: 28759049 DOI: 10.1038/nsmb.3444] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022]
Abstract
Toll-like receptor (TLR) signaling is a key innate immunity response to pathogens. Recruitment of signaling adapters such as MAL (TIRAP) and MyD88 to the TLRs requires Toll/interleukin-1 receptor (TIR)-domain interactions, which remain structurally elusive. Here we show that MAL TIR domains spontaneously and reversibly form filaments in vitro. They also form cofilaments with TLR4 TIR domains and induce formation of MyD88 assemblies. A 7-Å-resolution cryo-EM structure reveals a stable MAL protofilament consisting of two parallel strands of TIR-domain subunits in a BB-loop-mediated head-to-tail arrangement. Interface residues that are important for the interaction are conserved among different TIR domains. Although large filaments of TLR4, MAL or MyD88 are unlikely to form during cellular signaling, structure-guided mutagenesis, combined with in vivo interaction assays, demonstrated that the MAL interactions defined within the filament represent a template for a conserved mode of TIR-domain interaction involved in both TLR and interleukin-1 receptor signaling.
Collapse
|
14
|
Balaban S, Shearer RF, Lee LS, van Geldermalsen M, Schreuder M, Shtein HC, Cairns R, Thomas KC, Fazakerley DJ, Grewal T, Holst J, Saunders DN, Hoy AJ. Adipocyte lipolysis links obesity to breast cancer growth: adipocyte-derived fatty acids drive breast cancer cell proliferation and migration. Cancer Metab 2017; 5:1. [PMID: 28101337 PMCID: PMC5237166 DOI: 10.1186/s40170-016-0163-7] [Citation(s) in RCA: 295] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 12/26/2016] [Indexed: 12/31/2022] Open
Abstract
Background Obesity is associated with increased recurrence and reduced survival of breast cancer. Adipocytes constitute a significant component of breast tissue, yet their role in provisioning metabolic substrates to support breast cancer progression is poorly understood. Results Here, we show that co-culture of breast cancer cells with adipocytes revealed cancer cell-stimulated depletion of adipocyte triacylglycerol. Adipocyte-derived free fatty acids were transferred to breast cancer cells, driving fatty acid metabolism via increased CPT1A and electron transport chain complex protein levels, resulting in increased proliferation and migration. Notably, fatty acid transfer to breast cancer cells was enhanced from “obese” adipocytes, concomitant with increased stimulation of cancer cell proliferation and migration. This adipocyte-stimulated breast cancer cell proliferation was dependent on lipolytic processes since HSL/ATGL knockdown attenuated cancer cell responses. Conclusions These findings highlight a novel and potentially important role for adipocyte lipolysis in the provision of metabolic substrates to breast cancer cells, thereby supporting cancer progression. Electronic supplementary material The online version of this article (doi:10.1186/s40170-016-0163-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Seher Balaban
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, The Hub (D17), Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006 Australia
| | - Robert F Shearer
- Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW 2010 Australia
| | - Lisa S Lee
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, The Hub (D17), Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006 Australia
| | - Michelle van Geldermalsen
- Centenary Institute, The University of Sydney, Camperdown, NSW 2050 Australia ; Sydney Medical School, The University of Sydney, Camperdown, NSW 2006 Australia
| | - Mark Schreuder
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, The Hub (D17), Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006 Australia ; Faculty of Medicine, University of Utrecht, Utrecht, The Netherlands
| | - Harrison C Shtein
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, The Hub (D17), Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006 Australia
| | - Rose Cairns
- Faculty of Pharmacy, The University of Sydney, Camperdown, NSW 2006 Australia
| | - Kristen C Thomas
- School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006 Australia
| | - Daniel J Fazakerley
- School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006 Australia
| | - Thomas Grewal
- Faculty of Pharmacy, The University of Sydney, Camperdown, NSW 2006 Australia
| | - Jeff Holst
- Centenary Institute, The University of Sydney, Camperdown, NSW 2050 Australia ; Sydney Medical School, The University of Sydney, Camperdown, NSW 2006 Australia
| | - Darren N Saunders
- Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW 2010 Australia ; School of Medical Sciences, UNSW Australia, Sydney, NSW 2052 Australia
| | - Andrew J Hoy
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, The Hub (D17), Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006 Australia
| |
Collapse
|
15
|
Škalamera D, Dahmer-Heath M, Stevenson AJ, Pinto C, Shah ET, Daignault SM, Said NAB, Davis M, Haass NK, Williams ED, Hollier BG, Thompson EW, Gabrielli B, Gonda TJ. Genome-wide gain-of-function screen for genes that induce epithelial-to-mesenchymal transition in breast cancer. Oncotarget 2016; 7:61000-61020. [PMID: 27876705 PMCID: PMC5308632 DOI: 10.18632/oncotarget.11314] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/27/2016] [Indexed: 01/08/2023] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a developmental program that has been implicated in progression, metastasis and therapeutic resistance of some carcinomas. To identify genes whose overexpression drives EMT, we screened a lentiviral expression library of 17000 human open reading frames (ORFs) using high-content imaging to quantitate cytoplasmic vimentin. Hits capable of increasing vimentin in the mammary carcinoma-derived cell line MDA-MB-468 were confirmed in the non-tumorigenic breast-epithelial cell line MCF10A. When overexpressed in this model, they increased the rate of cell invasion through Matrigel™, induced mesenchymal marker expression and reduced expression of the epithelial marker E-cadherin. In gene-expression datasets derived from breast cancer patients, the expression of several novel genes correlated with expression of known EMT marker genes, indicating their in vivo relevance. As EMT-associated properties are thought to contribute in several ways to cancer progression, genes identified in this study may represent novel targets for anti-cancer therapy.
Collapse
Affiliation(s)
- Dubravka Škalamera
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
- Mater Medical Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Mareike Dahmer-Heath
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
- Mater Medical Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Alexander J. Stevenson
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
- Mater Medical Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Cletus Pinto
- St Vincent's Institute of Medical Research and University of Melbourne Department of Surgery, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Esha T. Shah
- Australian Prostate Cancer Research Centre-Queensland, Brisbane, QLD, Australia
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Translational Research Institute, Brisbane, QLD, Australia
| | - Sheena M. Daignault
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Nur Akmarina B.M. Said
- Monash Institute of Medical Research (now Hudson Institute of Medical Research), Monash University, Melbourne, VIC, Australia
- University of Malaya, Kuala Lumpur, Malaysia
| | - Melissa Davis
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Nikolas K. Haass
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Elizabeth D. Williams
- Australian Prostate Cancer Research Centre-Queensland, Brisbane, QLD, Australia
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Translational Research Institute, Brisbane, QLD, Australia
- Monash Institute of Medical Research (now Hudson Institute of Medical Research), Monash University, Melbourne, VIC, Australia
| | - Brett G. Hollier
- Australian Prostate Cancer Research Centre-Queensland, Brisbane, QLD, Australia
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Translational Research Institute, Brisbane, QLD, Australia
| | - Erik W. Thompson
- St Vincent's Institute of Medical Research and University of Melbourne Department of Surgery, St. Vincent's Hospital, Melbourne, VIC, Australia
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Translational Research Institute, Brisbane, QLD, Australia
| | - Brian Gabrielli
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
- Mater Medical Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Thomas J. Gonda
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
- School of Pharmacy, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
16
|
Patel J, Wong HY, Wang W, Alexis J, Shafiee A, Stevenson AJ, Gabrielli B, Fisk NM, Khosrotehrani K. Self-Renewal and High Proliferative Colony Forming Capacity of Late-Outgrowth Endothelial Progenitors Is Regulated by Cyclin-Dependent Kinase Inhibitors Driven by Notch Signaling. Stem Cells 2016; 34:902-12. [PMID: 26732848 DOI: 10.1002/stem.2262] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 11/10/2015] [Accepted: 11/30/2015] [Indexed: 11/10/2022]
Abstract
Since the discovery of endothelial colony forming cells (ECFC), there has been significant interest in their therapeutic potential to treat vascular injuries. ECFC cultures display significant heterogeneity and a hierarchy among cells able to give rise to high proliferative versus low proliferative colonies. Here we aimed to define molecularly this in vitro hierarchy. Based on flow cytometry, CD34 expression levels distinguished two populations. Only CD34 + ECFC had the capacity to reproduce high proliferative potential (HPP) colonies on replating, whereas CD34- ECFCs formed only small clusters. CD34 + ECFCs were the only ones to self-renew in stringent single-cell cultures and gave rise to both CD34 + and CD34- cells. Upon replating, CD34 + ECFCs were always found at the centre of HPP colonies and were more likely in G0/1 phase of cell cycling. Functionally, CD34 + ECFC were superior at restoring perfusion and better engrafted when injected into ischemic hind limbs. Transcriptomic analysis identified cyclin-dependent kinase (CDK) cell cycle inhibiting genes (p16, p21, and p57), the Notch signaling pathway (dll1, dll4, hes1, and hey1), and the endothelial cytokine il33 as highly expressed in CD34 + ECFC. Blocking the Notch pathway using a γ-secretase inhibitor (DAPT) led to reduced expression of cell cycle inhibitors, increased cell proliferation followed by a loss of self-renewal, and HPP colony formation capacity reflecting progenitor exhaustion. Similarly shRNA knockdown of p57 strongly affected self-renewal of ECFC colonies. ECFC hierarchy is defined by Notch signalling driving cell cycle regulators, progenitor quiescence and self-renewal potential.
Collapse
Affiliation(s)
- Jatin Patel
- UQ Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia
| | - Ho Yi Wong
- UQ Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia
| | - Weili Wang
- UQ Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia
| | - Josue Alexis
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Abbas Shafiee
- UQ Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia
| | - Alexander J Stevenson
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Brian Gabrielli
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Nicholas M Fisk
- UQ Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia.,Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Herston, Queensland, Australia
| | - Kiarash Khosrotehrani
- UQ Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia.,UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| |
Collapse
|
17
|
Gabrielli B, Bokhari F, Ranall MV, Oo ZY, Stevenson AJ, Wang W, Murrell M, Shaikh M, Fallaha S, Clarke D, Kelly M, Sedelies K, Christensen M, McKee S, Leggatt G, Leo P, Skalamera D, Soyer HP, Gonda TJ, McMillan NA. Aurora A Is Critical for Survival in HPV-Transformed Cervical Cancer. Mol Cancer Ther 2015; 14:2753-61. [DOI: 10.1158/1535-7163.mct-15-0506] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 10/04/2015] [Indexed: 11/16/2022]
|
18
|
Gagoski D, Polinkovsky ME, Mureev S, Kunert A, Johnston W, Gambin Y, Alexandrov K. Performance benchmarking of four cell-free protein expression systems. Biotechnol Bioeng 2015; 113:292-300. [PMID: 26301602 DOI: 10.1002/bit.25814] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/20/2015] [Accepted: 08/19/2015] [Indexed: 11/10/2022]
Abstract
Over the last half century, a range of cell-free protein expression systems based on pro- and eukaryotic organisms have been developed and have found a range of applications, from structural biology to directed protein evolution. While it is generally accepted that significant differences in performance among systems exist, there is a paucity of systematic experimental studies supporting this notion. Here, we took advantage of the species-independent translation initiation sequence to express and characterize 87 N-terminally GFP-tagged human cytosolic proteins of different sizes in E. coli, wheat germ (WGE), HeLa, and Leishmania-based (LTE) cell-free systems. Using a combination of single-molecule fluorescence spectroscopy, SDS-PAGE, and Western blot analysis, we assessed the expression yields, the fraction of full-length translation product, and aggregation propensity for each of these systems. Our results demonstrate that the E. coli system has the highest expression yields. However, we observe that high expression levels are accompanied by production of truncated species-particularly pronounced in the case of proteins larger than 70 kDa. Furthermore, proteins produced in the E. coli system display high aggregation propensity, with only 10% of tested proteins being produced in predominantly monodispersed form. The WGE system was the most productive among eukaryotic systems tested. Finally, HeLa and LTE show comparable protein yields that are considerably lower than the ones achieved in the E. coli and WGE systems. The protein products produced in the HeLa system display slightly higher integrity, whereas the LTE-produced proteins have the lowest aggregation propensity among the systems analyzed. The high quality of HeLa- and LTE-produced proteins enable their analysis without purification and make them suitable for analysis of multi-domain eukaryotic proteins.
Collapse
Affiliation(s)
- Dejan Gagoski
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, 4072, QLD, Australia
| | - Mark E Polinkovsky
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, 4072, QLD, Australia
| | - Sergey Mureev
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, 4072, QLD, Australia
| | - Anne Kunert
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, 4072, QLD, Australia
| | - Wayne Johnston
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, 4072, QLD, Australia
| | - Yann Gambin
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, 4072, QLD, Australia
| | - Kirill Alexandrov
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, 4072, QLD, Australia.
| |
Collapse
|
19
|
Ainger SA, Yong XL, Wong SS, Skalamera D, Gabrielli B, Leonard JH, Sturm RA. DCT protects human melanocytic cells from UVR and ROS damage and increases cell viability. Exp Dermatol 2015; 23:916-21. [PMID: 25346513 DOI: 10.1111/exd.12574] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2014] [Indexed: 12/25/2022]
Abstract
Dopachrome tautomerase (DCT) is involved in the formation of the photoprotective skin pigment eumelanin and has also been shown to have a role in response to apoptotic stimuli and oxidative stress. The effect of DCT on UVR DNA damage responses and survival pathways in human melanocytic cells was examined by knockdown experiments using melanoma cells, neonatal foreskin melanoblasts (MB) in monoculture and in co-culture with human keratinocytes. MB cell strains genotyped as either MC1R WT or MC1R RHC homozygotes, which are known to be deficient in DCT, were transduced with lentivirus vectors for either DCT knockdown or overexpression. We found melanoma cell survival was reduced by DCT depletion and by UVR over time. UVR-induced p53 and pp53-Ser15 levels were reduced with DCT depletion. Knockdown of DCT in MC1R WT and MC1R RHC MB cells reduced their survival after UVR exposure, whereas increased DCT protein levels enhanced survival. DCT depletion reduced p53 and pp53-Ser15 levels in WM266-4 melanoma and MC1R WT MB cells, while MC1R RHC MB cells displayed variable levels. Both MC1R WT and RHC genotypes of MB cells were responsive to UVR at 3 h with increases in both p53 and pp53-Ser15 proteins. MC1R WT MB cell strains in coculture with keratinocytes have an increased cell survival after UVR exposure when compared to those in monoculture, a protective effect which appears to be conferred by the keratinocytes.
Collapse
Affiliation(s)
- Stephen A Ainger
- Institute for Molecular Bioscience, Melanogenix Group, The University of Queensland, Brisbane, Qld, Australia
| | | | | | | | | | | | | |
Collapse
|
20
|
Gagoski D, Mureev S, Giles N, Johnston W, Dahmer-Heath M, Škalamera D, Gonda TJ, Alexandrov K. Gateway-compatible vectors for high-throughput protein expression in pro- and eukaryotic cell-free systems. J Biotechnol 2014; 195:1-7. [PMID: 25529348 DOI: 10.1016/j.jbiotec.2014.12.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 12/06/2014] [Accepted: 12/11/2014] [Indexed: 12/16/2022]
Abstract
Although numerous techniques for protein expression and production are available the pace of genome sequencing outstrips our ability to analyze the encoded proteins. To address this bottleneck, we have established a system for parallelized cloning, DNA production and cell-free expression of large numbers of proteins. This system is based on a suite of pCellFree Gateway destination vectors that utilize a Species Independent Translation Initiation Sequence (SITS) that mediates recombinant protein expression in any in vitro translation system. These vectors introduce C or N terminal EGFP and mCherry fluorescent and affinity tags, enabling direct analysis and purification of the expressed proteins. To maximize throughput and minimize the cost of protein production we combined Gateway cloning with Rolling Circle DNA Amplification. We demonstrate that as little as 0.1 ng of plasmid DNA is sufficient for template amplification and production of recombinant human protein in Leishmania tarentolae and Escherichia coli cell-free expression systems. Our experiments indicate that this approach can be applied to large gene libraries as it can be reliably performed in multi-well plates. The resulting protein expression pipeline provides a valuable new tool for applications of the post genomic era.
Collapse
Affiliation(s)
- Dejan Gagoski
- Institute for Molecular Bioscience, University of Queensland, Australia
| | - Sergey Mureev
- Institute for Molecular Bioscience, University of Queensland, Australia
| | - Nichole Giles
- Institute for Molecular Bioscience, University of Queensland, Australia
| | - Wayne Johnston
- Institute for Molecular Bioscience, University of Queensland, Australia
| | - Mareike Dahmer-Heath
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Dubravka Škalamera
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Thomas J Gonda
- School of Pharmacy, University of Queensland, Brisbane, Australia; University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Kirill Alexandrov
- Institute for Molecular Bioscience, University of Queensland, Australia.
| |
Collapse
|
21
|
Shearer RF, Saunders DN. Experimental design for stable genetic manipulation in mammalian cell lines: lentivirus and alternatives. Genes Cells 2014; 20:1-10. [PMID: 25307957 DOI: 10.1111/gtc.12183] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 08/04/2014] [Indexed: 01/22/2023]
Abstract
The use of third-generation lentiviral vectors is now commonplace in most areas of basic biology. These systems provide a fast, efficient means for modulating gene expression, but experimental design needs to be carefully considered to minimize potential artefacts arising from off-target effects and other confounding factors. This review offers a starting point for those new to lentiviral-based vector systems, addressing the main issues involved with the use of lentiviral systems in vitro and outlines considerations which should be taken into account during experimental design. Factors such as selecting an appropriate system and controls, and practical titration of viral transduction are important considerations for experimental design. We also briefly describe some of the more recent advances in genome editing technology. TALENs and CRISPRs offer an alternative to lentivirus, providing endogenous gene editing with reduced off-target effects often at the expense of efficiency.
Collapse
Affiliation(s)
- Robert F Shearer
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research and St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | | |
Collapse
|
22
|
Microscale acoustic disruption of mammalian cells for intracellular product release. J Biotechnol 2014; 184:146-53. [DOI: 10.1016/j.jbiotec.2014.04.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 03/21/2014] [Accepted: 04/28/2014] [Indexed: 11/16/2022]
|
23
|
Moon H, Lee CS, Inder KL, Sharma S, Choi E, Black DM, Lê Cao KA, Winterford C, Coward JI, Ling MT, Craik DJ, Parton RG, Russell PJ, Hill MM. PTRF/cavin-1 neutralizes non-caveolar caveolin-1 microdomains in prostate cancer. Oncogene 2014; 33:3561-70. [PMID: 23934189 DOI: 10.1038/onc.2013.315] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 06/08/2013] [Accepted: 06/11/2013] [Indexed: 12/15/2022]
Abstract
Caveolin-1 has a complex role in prostate cancer and has been suggested to be a potential biomarker and therapeutic target. As mature caveolin-1 resides in caveolae, invaginated lipid raft domains at the plasma membrane, caveolae have been suggested as a tumor-promoting signaling platform in prostate cancer. However, caveola formation requires both caveolin-1 and cavin-1 (also known as PTRF; polymerase I and transcript release factor). Here, we examined the expression of cavin-1 in prostate epithelia and stroma using tissue microarray including normal, non-malignant and malignant prostate tissues. We found that caveolin-1 was induced without the presence of cavin-1 in advanced prostate carcinoma, an expression pattern mirrored in the PC-3 cell line. In contrast, normal prostate epithelia expressed neither caveolin-1 nor cavin-1, while prostate stroma highly expressed both caveolin-1 and cavin-1. Utilizing PC-3 cells as a suitable model for caveolin-1-positive advanced prostate cancer, we found that cavin-1 expression in PC-3 cells inhibits anchorage-independent growth, and reduces in vivo tumor growth and metastasis in an orthotopic prostate cancer xenograft mouse model. The expression of α-smooth muscle actin in stroma along with interleukin-6 (IL-6) in cancer cells was also decreased in tumors of mice bearing PC-3-cavin-1 tumor cells. To determine whether cavin-1 acts by neutralizing caveolin-1, we expressed cavin-1 in caveolin-1-negative prostate cancer LNCaP and 22Rv1 cells. Caveolin-1 but not cavin-1 expression increased anchorage-independent growth in LNCaP and 22Rv1 cells. Cavin-1 co-expression reversed caveolin-1 effects in caveolin-1-positive LNCaP cells. Taken together, these results suggest that caveolin-1 in advanced prostate cancer is present outside of caveolae, because of the lack of cavin-1 expression. Cavin-1 expression attenuates the effects of non-caveolar caveolin-1 microdomains partly via reduced IL-6 microenvironmental function. With circulating caveolin-1 as a potential biomarker for advanced prostate cancer, identification of the molecular pathways affected by cavin-1 could provide novel therapeutic targets.
Collapse
Affiliation(s)
- H Moon
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - C S Lee
- 1] Discipline of Pathology, School of Medicine and Molecular Medicine Research Group, University of Western Sydney, Sydney, New South Wales, Australia [2] Department of Anatomical Pathology, Liverpool Hospital, Sydney, New South Wales, Australia
| | - K L Inder
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - S Sharma
- 1] Discipline of Pathology, School of Medicine and Molecular Medicine Research Group, University of Western Sydney, Sydney, New South Wales, Australia [2] Department of Anatomical Pathology, Liverpool Hospital, Sydney, New South Wales, Australia
| | - E Choi
- 1] The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia [2] School of Veterinary Science, The University of Queensland, Brisbane, Queensland, Australia
| | - D M Black
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - K-A Lê Cao
- Queensland Facility for Advanced Bioinformatics, The University of Queensland, Brisbane, Queensland, Australia
| | - C Winterford
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - J I Coward
- Mater Research, Translational Research Institute, Brisbane, Queensland, Australia
| | - M T Ling
- Australian Prostate Cancer Research Centre-Queensland and Institute for Biomedical Health & Innovation, Queensland University of Technology, Translational Research Institute, Brisbane, Queensland, Australia
| | - D J Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - R G Parton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - P J Russell
- Australian Prostate Cancer Research Centre-Queensland and Institute for Biomedical Health & Innovation, Queensland University of Technology, Translational Research Institute, Brisbane, Queensland, Australia
| | - M M Hill
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
24
|
Sierecki E, Stevers LM, Giles N, Polinkovsky ME, Moustaqil M, Mureev S, Johnston WA, Dahmer-Heath M, Skalamera D, Gonda TJ, Gabrielli B, Collins BM, Alexandrov K, Gambin Y. Rapid mapping of interactions between Human SNX-BAR proteins measured in vitro by AlphaScreen and single-molecule spectroscopy. Mol Cell Proteomics 2014; 13:2233-45. [PMID: 24866125 DOI: 10.1074/mcp.m113.037275] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein dimerization and oligomerization is commonly used by nature to increase the structural and functional complexity of proteins. Regulated protein assembly is essential to transfer information in signaling, transcriptional, and membrane trafficking events. Here we show that a combination of cell-free protein expression, a proximity based interaction assay (AlphaScreen), and single-molecule fluorescence allow rapid mapping of homo- and hetero-oligomerization of proteins. We have applied this approach to the family of BAR domain-containing sorting nexin (SNX-BAR) proteins, which are essential regulators of membrane trafficking and remodeling in all eukaryotes. Dimerization of BAR domains is essential for creating a concave structure capable of sensing and inducing membrane curvature. We have systematically mapped 144 pairwise interactions between the human SNX-BAR proteins and generated an interaction matrix of preferred dimerization partners for each family member. We find that while nine SNX-BAR proteins are able to form homo-dimers, several including the retromer-associated SNX1, SNX2, and SNX5 require heteromeric interactions for dimerization. SNX2, SNX4, SNX6, and SNX8 show a promiscuous ability to bind other SNX-BAR proteins and we also observe a novel interaction with the SNX3 protein which lacks the BAR domain structure.
Collapse
Affiliation(s)
- Emma Sierecki
- From the ‡Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| | - Loes M Stevers
- From the ‡Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| | - Nichole Giles
- From the ‡Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| | - Mark E Polinkovsky
- From the ‡Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| | - Mehdi Moustaqil
- From the ‡Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| | - Sergey Mureev
- From the ‡Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| | - Wayne A Johnston
- From the ‡Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| | - Mareike Dahmer-Heath
- §University of Queensland, Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Dubravka Skalamera
- §University of Queensland, Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Thomas J Gonda
- ¶School of Pharmacy, The University of Queensland, Brisbane, Australia
| | - Brian Gabrielli
- §University of Queensland, Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Brett M Collins
- From the ‡Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| | - Kirill Alexandrov
- From the ‡Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia;
| | - Yann Gambin
- From the ‡Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| |
Collapse
|
25
|
Arnoldo A, Kittanakom S, Heisler LE, Mak AB, Shukalyuk AI, Torti D, Moffat J, Giaever G, Nislow C. A genome scale overexpression screen to reveal drug activity in human cells. Genome Med 2014; 6:32. [PMID: 24944581 PMCID: PMC4062067 DOI: 10.1186/gm549] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 04/22/2014] [Indexed: 02/08/2023] Open
Abstract
Target identification is a critical step in the lengthy and expensive process of drug development. Here, we describe a genome-wide screening platform that uses systematic overexpression of pooled human ORFs to understand drug mode-of-action and resistance mechanisms. We first calibrated our screen with the well-characterized drug methotrexate. We then identified new genes involved in the bioactivity of diverse drugs including antineoplastic agents and biologically active molecules. Finally, we focused on the transcription factor RHOXF2 whose overexpression conferred resistance to DNA damaging agents. This approach represents an orthogonal method for functional screening and, to our knowledge, has never been reported before.
Collapse
Affiliation(s)
- Anthony Arnoldo
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 3E1, Canada ; Banting and Best Department of Medical Research, University of Toronto, Toronto, M5S 3E1, Canada ; Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Saranya Kittanakom
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 3E1, Canada ; Banting and Best Department of Medical Research, University of Toronto, Toronto, M5S 3E1, Canada ; Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Lawrence E Heisler
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 3E1, Canada ; Banting and Best Department of Medical Research, University of Toronto, Toronto, M5S 3E1, Canada ; Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada ; Donnelly Sequencing Center, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Anthony B Mak
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 3E1, Canada ; Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Andrey I Shukalyuk
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College Street, Toronto M5S 3E3, Canada
| | - Dax Torti
- Donnelly Sequencing Center, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Jason Moffat
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 3E1, Canada ; Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Guri Giaever
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 3E1, Canada ; Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada ; Department of Pharmaceutical Sciences, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada ; Department of Pharmaceutical Sciences, University of British Columbia, 6619-2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Corey Nislow
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 3E1, Canada ; Banting and Best Department of Medical Research, University of Toronto, Toronto, M5S 3E1, Canada ; Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada ; Donnelly Sequencing Center, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada ; Department of Pharmaceutical Sciences, University of British Columbia, 6619-2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
26
|
Bersten DC, Bruning JB, Peet DJ, Whitelaw ML. Human variants in the neuronal basic helix-loop-helix/Per-Arnt-Sim (bHLH/PAS) transcription factor complex NPAS4/ARNT2 disrupt function. PLoS One 2014; 9:e85768. [PMID: 24465693 PMCID: PMC3894988 DOI: 10.1371/journal.pone.0085768] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 12/06/2013] [Indexed: 11/25/2022] Open
Abstract
Neuronal Per-Arnt-Sim homology (PAS) Factor 4 (NPAS4) is a neuronal activity-dependent transcription factor which heterodimerises with ARNT2 to regulate genes involved in inhibitory synapse formation. NPAS4 functions to maintain excitatory/inhibitory balance in neurons, while mouse models have shown it to play roles in memory formation, social interaction and neurodegeneration. NPAS4 has therefore been implicated in a number of neuropsychiatric or neurodegenerative diseases which are underpinned by defects in excitatory/inhibitory balance. Here we have explored a broad set of non-synonymous human variants in NPAS4 and ARNT2 for disruption of NPAS4 function. We found two variants in NPAS4 (F147S and E257K) and two variants in ARNT2 (R46W and R107H) which significantly reduced transcriptional activity of the heterodimer on a luciferase reporter gene. Furthermore, we found that NPAS4.F147S was unable to activate expression of the NPAS4 target gene BDNF due to reduced dimerisation with ARNT2. Homology modelling predicts F147 in NPAS4 to lie at the dimer interface, where it appears to directly contribute to protein/protein interaction. We also found that reduced transcriptional activation by ARNT2 R46W was due to disruption of nuclear localisation. These results provide insight into the mechanisms of NPAS4/ARNT dimerisation and transcriptional activation and have potential implications for cognitive phenotypic variation and diseases such as autism, schizophrenia and dementia.
Collapse
Affiliation(s)
- David C. Bersten
- School of Molecular and Biomedical Science (Biochemistry), and Australian Research Council Special Research Centre for the Molecular Genetics of Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - John B. Bruning
- School of Molecular and Biomedical Science (Biochemistry), and Australian Research Council Special Research Centre for the Molecular Genetics of Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - Daniel J. Peet
- School of Molecular and Biomedical Science (Biochemistry), and Australian Research Council Special Research Centre for the Molecular Genetics of Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - Murray L. Whitelaw
- School of Molecular and Biomedical Science (Biochemistry), and Australian Research Council Special Research Centre for the Molecular Genetics of Development, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
27
|
Hugo HJ, Pereira L, Suryadinata R, Drabsch Y, Gonda TJ, Gunasinghe NPAD, Pinto C, Soo ETL, van Denderen BJW, Hill P, Ramsay RG, Sarcevic B, Newgreen DF, Thompson EW. Direct repression of MYB by ZEB1 suppresses proliferation and epithelial gene expression during epithelial-to-mesenchymal transition of breast cancer cells. Breast Cancer Res 2013; 15:R113. [PMID: 24283570 PMCID: PMC3979034 DOI: 10.1186/bcr3580] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 10/31/2013] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Epithelial-to-mesenchymal transition (EMT) promotes cell migration and is important in metastasis. Cellular proliferation is often downregulated during EMT, and the reverse transition (MET) in metastases appears to be required for restoration of proliferation in secondary tumors. We studied the interplay between EMT and proliferation control by MYB in breast cancer cells. METHODS MYB, ZEB1, and CDH1 expression levels were manipulated by lentiviral small-hairpin RNA (shRNA)-mediated knockdown/overexpression, and verified with Western blotting, immunocytochemistry, and qRT-PCR. Proliferation was assessed with bromodeoxyuridine pulse labeling and flow cytometry, and sulforhodamine B assays. EMT was induced with epidermal growth factor for 9 days or by exposure to hypoxia (1% oxygen) for up to 5 days, and assessed with qRT-PCR, cell morphology, and colony morphology. Protein expression in human breast cancers was assessed with immunohistochemistry. ZEB1-MYB promoter binding and repression were determined with Chromatin Immunoprecipitation Assay and a luciferase reporter assay, respectively. Student paired t tests, Mann-Whitney, and repeated measures two-way ANOVA tests determined statistical significance (P < 0.05). RESULTS Parental PMC42-ET cells displayed higher expression of ZEB1 and lower expression of MYB than did the PMC42-LA epithelial variant. Knockdown of ZEB1 in PMC42-ET and MDA-MB-231 cells caused increased expression of MYB and a transition to a more epithelial phenotype, which in PMC42-ET cells was coupled with increased proliferation. Indeed, we observed an inverse relation between MYB and ZEB1 expression in two in vitro EMT cell models, in matched human breast tumors and lymph node metastases, and in human breast cancer cell lines. Knockdown of MYB in PMC42-LA cells (MYBsh-LA) led to morphologic changes and protein expression consistent with an EMT. ZEB1 expression was raised in MYBsh-LA cells and significantly repressed in MYB-overexpressing MDA-MB-231 cells, which also showed reduced random migration and a shift from mesenchymal to epithelial colony morphology in two dimensional monolayer cultures. Finally, we detected binding of ZEB1 to MYB promoter in PMC42-ET cells, and ZEB1 overexpression repressed MYB promoter activity. CONCLUSIONS This work identifies ZEB1 as a transcriptional repressor of MYB and suggests a reciprocal MYB-ZEB1 repressive relation, providing a mechanism through which proliferation and the epithelial phenotype may be coordinately modulated in breast cancer cells.
Collapse
|
28
|
Zhao L, Ye P, Gonda TJ. The MYB proto-oncogene suppresses monocytic differentiation of acute myeloid leukemia cells via transcriptional activation of its target gene GFI1. Oncogene 2013; 33:4442-9. [PMID: 24121275 DOI: 10.1038/onc.2013.419] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/14/2013] [Accepted: 09/02/2013] [Indexed: 12/20/2022]
Abstract
The MYB gene is a master regulator of hematopoiesis and contributes to leukemogenesis in several species including humans. Although it is clear that MYB can promote proliferation, suppress apoptosis and block differentiation, the identities of the MYB target genes that mediate these effects have only been partially elucidated. Several studies, including our own, have collectively identified substantial numbers of MYB target genes, including candidates for each of these activities; however, functional validation, particularly in the case of differentiation suppression, has lagged well behind. Here we show that GFI1, which encodes an important regulator of hematopoietic stem cell (HSC) function and granulocytic differentiation, is a direct target of MYB in myeloid leukemia cells. Chromatin immunoprecipitation and reporter studies identified a functional MYB-binding site in the promoter region of GFI, whereas ectopic expression and small hairpin RNA-mediated knockdown of MYB resulted in concomitant increases and decreases, respectively, in GFI1 expression. We also demonstrate that GFI1, like MYB, can block the induced monocytic differentiation of a human acute myeloid leukemia cell line, and most importantly, that GFI1 is essential for MYB's ability to block monocytic differentiation. Thus, we have identified a target of MYB that is a likely mediator of its myeloid differentiation-blocking activity, and which may also be involved in MYB's activities in regulating normal HSC function and myeloid differentiation.
Collapse
Affiliation(s)
- L Zhao
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - P Ye
- 1] The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia [2] School of Pharmacy, The University of Queensland, Pharmacy Australia Centre of Excellence, Brisbane, Queensland, Australia
| | - T J Gonda
- 1] The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia [2] School of Pharmacy, The University of Queensland, Pharmacy Australia Centre of Excellence, Brisbane, Queensland, Australia
| |
Collapse
|
29
|
Stone JD, Kranz DM. Role of T cell receptor affinity in the efficacy and specificity of adoptive T cell therapies. Front Immunol 2013; 4:244. [PMID: 23970885 PMCID: PMC3748443 DOI: 10.3389/fimmu.2013.00244] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/05/2013] [Indexed: 01/09/2023] Open
Abstract
Over the last several years, there has been considerable progress in the treatment of cancer using gene modified adoptive T cell therapies. Two approaches have been used, one involving the introduction of a conventional αβ T cell receptor (TCR) against a pepMHC cancer antigen, and the second involving introduction of a chimeric antigen receptor (CAR) consisting of a single-chain antibody as an Fv fragment linked to transmembrane and signaling domains. In this review, we focus on one aspect of TCR-mediated adoptive T cell therapies, the impact of the affinity of the αβ TCR for the pepMHC cancer antigen on both efficacy and specificity. We discuss the advantages of higher-affinity TCRs in mediating potent activity of CD4 T cells. This is balanced with the potential disadvantage of higher-affinity TCRs in mediating greater self-reactivity against a wider range of structurally similar antigenic peptides, especially in synergy with the CD8 co-receptor. Both TCR affinity and target selection will influence potential safety issues. We suggest pre-clinical strategies that might be used to examine each TCR for possible on-target and off-target side effects due to self-reactivities, and to adjust TCR affinities accordingly.
Collapse
Affiliation(s)
- Jennifer D Stone
- Department of Biochemistry, University of Illinois , Urbana, IL , USA
| | | |
Collapse
|
30
|
Developmental stage dependent neural stem cells sensitivity to methylmercury chloride on different biofunctional surfaces. Toxicol In Vitro 2013; 28:76-87. [PMID: 23872426 DOI: 10.1016/j.tiv.2013.06.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 06/10/2013] [Accepted: 06/26/2013] [Indexed: 12/29/2022]
Abstract
Sensitivity of neural stem cells viability, proliferation and differentiation upon exposure to methylmercury chloride (MeHgCl) was investigated on different types of biofunctional surfaces. Patterns of biodomains created by microprinting/microspotting of poly-l-lysine or extracellular matrix proteins (fibronectin and vitronectin) allowed for non-specific electrostatic or specific, receptor mediated interactions, respectively, between stem cells and the surface. The neural stem cell line HUCB-NSC has been previously shown to be susceptible to MeHgCl in developmentally dependent manner. Here we demonstrated that developmental sensitivity of HUCB-NSC to MeHgCl depends upon the type of adhesive biomolecules and the geometry of biodomains. Proliferation of HUCB-NSC was diminished in time and MeHgCl concentration dependent manner. In addition, the response to MeHgCl was found to be cell-type dependent. Undifferentiated cells were the most sensitive independently of the type of bioactive domain. Significant decrease of GFAP+ cells was detected among cells growing on poly-l-lysine, while on fibronectin and vitronectin, this effect was observed only in the highest (1μM) concentration of MeHgCl. β-Tubulin III expressing cells were most sensitive on fibronectin domains. In addition, limited bioactive domains to μm in size, as compared to non-patterned larger area of the same adhesive substrate, exerted protective role. Thus, the surface area and type of cell/biofunctional surface interaction exerted significant influence on developmental stage and cell-type specific response of HUCB-NSC to MeHgCl.
Collapse
|
31
|
Zhang H, Wang L, Hunter D, Voogd C, Joyce N, Davies K. A Narcissus mosaic viral vector system for protein expression and flavonoid production. PLANT METHODS 2013; 9:28. [PMID: 23849589 PMCID: PMC3728148 DOI: 10.1186/1746-4811-9-28] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 07/07/2013] [Indexed: 05/24/2023]
Abstract
BACKGROUND With the explosive numbers of sequences generated by next generation sequencing, the demand for high throughput screening to understand gene function has grown. Plant viral vectors have been widely used as tools in down-regulating plant gene expression. However, plant viral vectors can also express proteins in a very efficient manner and, therefore, can also serve as a valuable tool for characterizing proteins and their functions in metabolic pathways in planta. RESULTS In this study, we have developed a Gateway®-based high throughput viral vector cloning system from Narcissus Mosaic Virus (NMV). Using the reporter genes of GFP and GUS, and the plant genes PAP1 (an R2R3 MYB which activates the anthocyanin pathway) and selenium-binding protein 1 (SeBP), we show that NMV vectors and the model plant Nicotiana benthamiana can be used for efficient protein expression, protein subcellular localization and secondary metabolite production. CONCLUSIONS Our results suggest that not only can the plant viral vector system be employed for protein work but also can potentially be amenable to producing valuable secondary metabolites on a large scale, as the system does not require plant regeneration from seed or calli, which are stages where certain secondary metabolites can interfere with development.
Collapse
Affiliation(s)
- Huaibi Zhang
- The New Zealand Institute for Plant & Food Research Limited (PFR), Private Bag 11600 Palmerston North, New Zealand
| | - Lei Wang
- The New Zealand Institute for Plant & Food Research Limited (PFR), Private Bag 11600 Palmerston North, New Zealand
| | - Donald Hunter
- The New Zealand Institute for Plant & Food Research Limited (PFR), Private Bag 11600 Palmerston North, New Zealand
| | - Charlotte Voogd
- PFR, Private Bag Private Bag 92169, Auckland 1142 New Zealand
| | - Nigel Joyce
- PFR, Private Bag 4704 Christchurch, New Zealand
| | - Kevin Davies
- The New Zealand Institute for Plant & Food Research Limited (PFR), Private Bag 11600 Palmerston North, New Zealand
| |
Collapse
|
32
|
Festa F, Steel J, Bian X, Labaer J. High-throughput cloning and expression library creation for functional proteomics. Proteomics 2013; 13:1381-99. [PMID: 23457047 DOI: 10.1002/pmic.201200456] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 01/11/2013] [Accepted: 01/31/2013] [Indexed: 12/11/2022]
Abstract
The study of protein function usually requires the use of a cloned version of the gene for protein expression and functional assays. This strategy is particularly important when the information available regarding function is limited. The functional characterization of the thousands of newly identified proteins revealed by genomics requires faster methods than traditional single-gene experiments, creating the need for fast, flexible, and reliable cloning systems. These collections of ORF clones can be coupled with high-throughput proteomics platforms, such as protein microarrays and cell-based assays, to answer biological questions. In this tutorial, we provide the background for DNA cloning, discuss the major high-throughput cloning systems (Gateway® Technology, Flexi® Vector Systems, and Creator(TM) DNA Cloning System) and compare them side-by-side. We also report an example of high-throughput cloning study and its application in functional proteomics. This tutorial is part of the International Proteomics Tutorial Programme (IPTP12).
Collapse
Affiliation(s)
- Fernanda Festa
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85287-6401, USA
| | | | | | | |
Collapse
|
33
|
Liu Y, Yang X, Guo C, Nie P, Liu Y, Ma J. Essential role of MFG-E8 for phagocytic properties of microglial cells. PLoS One 2013; 8:e55754. [PMID: 23405209 PMCID: PMC3565973 DOI: 10.1371/journal.pone.0055754] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 12/31/2012] [Indexed: 12/20/2022] Open
Abstract
Milk fat globule factor-E8 (MFG-E8) has been regarded as a key factor involved in the phagocytosis of apoptotic cells. We induced a lentivirus into the microglial cells for the augmentation or abrogation of MFG-E8 expression in mouse microglial cells, and investigated phagocytosis of phosphatidylserine tagged human red blood cells (hRBCs) in co-cultures. Increased MFG-E8 levels were associated with a significant increase in phagocytic activity compared to the controls. Conversely, phagocytosis dramitically decreased due to the abrogation of MFG-E8. In addition, the expression of the inflammatory cytokines, TNF-α and IL-1β, also increased or decreased in the microglial cells with the augmentation or abrogation of MFG-E8, respectively. Our findings indicate that the enhanced expression of MFG-E8 could increase phagocytosis of apoptotic cells; conversely, the rate of phagocytosis and the expression of inflammatory cytokines decreased when MFG-E8 expression was knocked down. Our results confirm that MFG-E8 plays an important role in phagocytosis, and possibly serves as an essential signal molecule for microglial cells.
Collapse
Affiliation(s)
- Yong Liu
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University, Chongqing, China.
| | | | | | | | | | | |
Collapse
|
34
|
Guo J, Li D, Bai S, Xu T, Zhou Z, Zhang Y. Detecting DNA synthesis of neointimal formation after catheter balloon injury in GK and in Wistar rats: using 5-ethynyl-2'-deoxyuridine. Cardiovasc Diabetol 2012; 11:150. [PMID: 23237526 PMCID: PMC3537571 DOI: 10.1186/1475-2840-11-150] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 10/13/2012] [Indexed: 01/19/2023] Open
Abstract
Background Neointimal formation plays an important role in the pathogenesis of coronary restenosis after percutaneous coronary intervention (PCI), especially in patients with diabetes mellitus. Recently, some studies have shown that 5-ethynyl-2'-deoxyuridine (EdU) incorporation can serve as a novel alternative to the 5-bromo-2'-deoxyuridine (BrdU) antibody detection method for detection of DNA synthesis in regenerating avian cochlea, chick embryo and the adult nervous system. However, few studies have been performed to assess the suitability of EdU for detecting DNA synthesis in vascular neointima. Methods The carotid artery balloon injury model was established in Goto-Kakizaki (GK) and Wistar rats. A Cell-LightTM EdU Kit was used to detect EdU-labeled cell nuclei of common carotid arteries at day 7 after catheter balloon injury. Different methods of injecting EdU were tested. The protein levels of proliferating cell nuclear antigen (PCNA) and p-Akt (Ser473), as well as the mRNA levels of PCNA were evaluated by Western blotting and quantitative real-time PCR (qRT-PCR), respectively. Immunohistochemical staining was also employed to visualize PCNA-positive cells. Results At day 7 after catheter balloon injury, far more EdU-positive and PCNA-positive cells were observed in GK rats. When comparing groups that received different EdU doses, it was found that the percentage of EdU-positive cells at a dose of 100 mg/kg body weight was than at doses of 25 mg/kg and 50 mg/kg. The number of positive cells was significantly higher in the repeated injection group compared to the single injection group. Further, after balloon injury DNA synthesis in GK rats was more notable than in Wistar rats. Neointimal formation in GK rats was more obvious than in Wistar rats. The protein levels of PCNA and p-Akt (Ser473) and the mRNA levels of PCNA were increased in injured rats as compared to uninjured rats, and were significantly higher in GK rats than in Wistar rats. Conclusion By intraperitoneal injections of EdU at a dose of 100 mg/kg three times, EdU incorporation can detect carotid arterial DNA synthesis caused by neointimal formation in GK rats and Wistar rats at day 7 after balloon injury by the EdU click reaction quickly and effectively. Moreover, more obvious DNA synthesis in the vascular neointima could be observed in GK rats than in Wistar rats.
Collapse
Affiliation(s)
- Jingsheng Guo
- Institute of Cardiovascular Disease Research, Xuzhou Medical College, 84 West Huaihai Road, Xuzhou, Jiangsu, Peoples Republic of China
| | | | | | | | | | | |
Collapse
|
35
|
Generation of a genome scale lentiviral vector library for EF1α promoter-driven expression of human ORFs and identification of human genes affecting viral titer. PLoS One 2012; 7:e51733. [PMID: 23251614 PMCID: PMC3520899 DOI: 10.1371/journal.pone.0051733] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 11/06/2012] [Indexed: 01/12/2023] Open
Abstract
The bottleneck in elucidating gene function through high-throughput gain-of-function genome screening is the limited availability of comprehensive libraries for gene overexpression. Lentiviral vectors are the most versatile and widely used vehicles for gene expression in mammalian cells. Lentiviral supernatant libraries for genome screening are commonly generated in the HEK293T cell line, yet very little is known about the effect of introduced sequences on the produced viral titer, which we have shown to be gene dependent. We have generated an arrayed lentiviral vector library for the expression of 17,030 human proteins by using the GATEWAY® cloning system to transfer ORFs from the Mammalian Gene Collection into an EF1alpha promoter-dependent lentiviral expression vector. This promoter was chosen instead of the more potent and widely used CMV promoter, because it is less prone to silencing and provides more stable long term expression. The arrayed lentiviral clones were used to generate viral supernatant by packaging in the HEK293T cell line. The efficiency of transfection and virus production was estimated by measuring the fluorescence of IRES driven GFP, co-expressed with the ORFs. More than 90% of cloned ORFs produced sufficient virus for downstream screening applications. We identified genes which consistently produced very high or very low viral titer. Supernatants from select clones that were either high or low virus producers were tested on a range of cell lines. Some of the low virus producers, including two previously uncharacterized proteins were cytotoxic to HEK293T cells. The library we have constructed presents a powerful resource for high-throughput gain-of-function screening of the human genome and drug-target discovery. Identification of human genes that affect lentivirus production may lead to improved technology for gene expression using lentiviral vectors.
Collapse
|
36
|
Maier CJ, Maier RH, Virok DP, Maass M, Hintner H, Bauer JW, Onder K. Construction of a highly flexible and comprehensive gene collection representing the ORFeome of the human pathogen Chlamydia pneumoniae. BMC Genomics 2012; 13:632. [PMID: 23157390 PMCID: PMC3534531 DOI: 10.1186/1471-2164-13-632] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 11/11/2012] [Indexed: 12/02/2022] Open
Abstract
Background The Gram-negative bacterium Chlamydia pneumoniae (Cpn) is the leading intracellular human pathogen responsible for respiratory infections such as pneumonia and bronchitis. Basic and applied research in pathogen biology, especially the elaboration of new mechanism-based anti-pathogen strategies, target discovery and drug development, rely heavily on the availability of the entire set of pathogen open reading frames, the ORFeome. The ORFeome of Cpn will enable genome- and proteome-wide systematic analysis of Cpn, which will improve our understanding of the molecular networks and mechanisms underlying and governing its pathogenesis. Results Here we report the construction of a comprehensive gene collection covering 98.5% of the 1052 predicted and verified ORFs of Cpn (Chlamydia pneumoniae strain CWL029) in Gateway® ‘entry’ vectors. Based on genomic DNA isolated from the vascular chlamydial strain CV-6, we constructed an ORFeome library that contains 869 unique Gateway® entry clones (83% coverage) and an additional 168 PCR-verified ‘pooled’ entry clones, reaching an overall coverage of ~98.5% of the predicted CWL029 ORFs. The high quality of the ORFeome library was verified by PCR-gel electrophoresis and DNA sequencing, and its functionality was demonstrated by expressing panels of recombinant proteins in Escherichia coli and by genome-wide protein interaction analysis for a test set of three Cpn virulence factors in a yeast 2-hybrid system. The ORFeome is available in different configurations of resource stocks, PCR-products, purified plasmid DNA, and living cultures of E. coli harboring the desired entry clone or pooled entry clones. All resources are available in 96-well microtiterplates. Conclusion This first ORFeome library for Cpn provides an essential new tool for this important pathogen. The high coverage of entry clones will enable a systems biology approach for Cpn or host–pathogen analysis. The high yield of recombinant proteins and the promising interactors for Cpn virulence factors described here demonstrate the possibilities for proteome-wide studies.
Collapse
Affiliation(s)
- Christina J Maier
- Department of Dermatology, Paracelsus Medical University, Salzburg, Austria
| | | | | | | | | | | | | |
Collapse
|
37
|
Heynen-Genel S, Pache L, Chanda SK, Rosen J. Functional genomic and high-content screening for target discovery and deconvolution. Expert Opin Drug Discov 2012; 7:955-68. [PMID: 22860749 DOI: 10.1517/17460441.2012.711311] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Functional genomic screens apply knowledge gained from the sequencing of the human genome toward rapid methods of identifying genes involved in cellular function based on a specific phenotype. This approach has been made possible through advances in both molecular biology and automation. The utility of this approach has been further enhanced through the application of image-based high-content screening: an automated microscopy and quantitative image analysis platform. These approaches can significantly enhance the acquisition of novel targets for drug discovery. AREAS COVERED Both the utility and potential issues associated with functional genomic screening approaches are discussed in this review, along with examples that illustrate both. The considerations for high-content screening applied to functional genomics are also presented. EXPERT OPINION Functional genomic screening and high-content screening are extremely useful in the identification of new drug targets. However, the technical, experimental, and computational parameters have an enormous influence on the results. Thus, although new targets are identified, caution should be applied to the interpretation of screening data in isolation. Genomic screens should be viewed as an integral component of a target identification campaign that requires both the acquisition of orthogonal data, as well as a rigorous validation strategy.
Collapse
|
38
|
Tralau T, Luch A. Drug-mediated toxicity: illuminating the ‘bad’ in the test tube by means of cellular assays? Trends Pharmacol Sci 2012; 33:353-64. [DOI: 10.1016/j.tips.2012.03.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 03/12/2012] [Accepted: 03/28/2012] [Indexed: 12/19/2022]
|
39
|
Ranga A, Lutolf MP. High-throughput approaches for the analysis of extrinsic regulators of stem cell fate. Curr Opin Cell Biol 2012; 24:236-44. [DOI: 10.1016/j.ceb.2012.01.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 01/12/2012] [Indexed: 01/10/2023]
|
40
|
Hossain MS, Gresock J, Edmonds Y, Helm R, Potts M, Ramakrishnan N. Connecting the dots between PubMed abstracts. PLoS One 2012; 7:e29509. [PMID: 22235301 PMCID: PMC3250456 DOI: 10.1371/journal.pone.0029509] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 11/29/2011] [Indexed: 11/23/2022] Open
Abstract
Background There are now a multitude of articles published in a diversity of journals providing information about genes, proteins, pathways, and diseases. Each article investigates subsets of a biological process, but to gain insight into the functioning of a system as a whole, we must integrate information from multiple publications. Particularly, unraveling relationships between extra-cellular inputs and downstream molecular response mechanisms requires integrating conclusions from diverse publications. Methodology We present an automated approach to biological knowledge discovery from PubMed abstracts, suitable for “connecting the dots” across the literature. We describe a storytelling algorithm that, given a start and end publication, typically with little or no overlap in content, identifies a chain of intermediate publications from one to the other, such that neighboring publications have significant content similarity. The quality of discovered stories is measured using local criteria such as the size of supporting neighborhoods for each link and the strength of individual links connecting publications, as well as global metrics of dispersion. To ensure that the story stays coherent as it meanders from one publication to another, we demonstrate the design of novel coherence and overlap filters for use as post-processing steps. Conclusions We demonstrate the application of our storytelling algorithm to three case studies: i) a many-one study exploring relationships between multiple cellular inputs and a molecule responsible for cell-fate decisions, ii) a many-many study exploring the relationships between multiple cytokines and multiple downstream transcription factors, and iii) a one-to-one study to showcase the ability to recover a cancer related association, viz. the Warburg effect, from past literature. The storytelling pipeline helps narrow down a scientist's focus from several hundreds of thousands of relevant documents to only around a hundred stories. We argue that our approach can serve as a valuable discovery aid for hypothesis generation and connection exploration in large unstructured biological knowledge bases.
Collapse
Affiliation(s)
- M Shahriar Hossain
- Department of Computer Science, Virginia Tech, Blacksburg, Virginia, United States of America.
| | | | | | | | | | | |
Collapse
|