1
|
Zhu X, Chen J, Liao J, Wang M, Long Y, Liu M, Zhang Y, Wang HH. Functionalized Multichannel Fluorescence-Encoded Nanosystem on Erythrocyte-Coated Nanoparticles for Precise Cancer Subtype Discrimination. NANO LETTERS 2025; 25:426-433. [PMID: 39690886 DOI: 10.1021/acs.nanolett.4c05208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Rapid and precise cancer subtype discrimination is essential for personalized oncology. Conventional diagnostic methods often lack sufficient accuracy and speed. Here, we introduce a multichannel fluorescence-encoded nanosystem based on erythrocyte-coated polydopamine nanoparticles (PDA@EM), functionalized with multiple resurfaced fluorescent proteins. The fluorescence of these proteins is initially quenched by PDA@EM and restored upon cell addition. This multichannel fluorescence-encoded nanosystem enables highly sensitive "turn-on" fluorescence profiling of cancer cells within 30 min, achieving 100% accuracy in distinguishing various proteins and classifying a wide range of cancer cell lines, including subtypes of oral squamous cell carcinoma (OSCC). Notably, it offers rapid, label-free diagnostics of OSCC malignancy from clinical samples postsurgery. This capability was validated through histopathological and proteomic analyses, which identified protein signatures associated with tumor progression and immune suppression. Overall, our multichannel nanosensor represents an advanced molecular diagnostics platform, paving the way for personalized cancer treatment in clinical oncology.
Collapse
Affiliation(s)
- Xiaohua Zhu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China
| | - Jiali Chen
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China
| | - Junyu Liao
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China
| | - Meixia Wang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Ying Long
- The Affiliated Cancer Hospital of Xiangya School of Medicine/Hunan Cancer Hospital, Central South University, Changsha 410013, China
| | - Meiling Liu
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China
| | - Youyu Zhang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P. R. China
| | - Hong-Hui Wang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| |
Collapse
|
2
|
Lévrier A, Capin J, Mayonove P, Karpathakis II, Voyvodic P, DeVisch A, Zuniga A, Cohen-Gonsaud M, Cabantous S, Noireaux V, Bonnet J. Split Reporters Facilitate Monitoring of Gene Expression and Peptide Production in Linear Cell-Free Transcription-Translation Systems. ACS Synth Biol 2024; 13:3119-3127. [PMID: 39292739 DOI: 10.1021/acssynbio.4c00353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Cell-free transcription-translation (TXTL) systems expressing genes from linear dsDNA enable the rapid prototyping of genetic devices while avoiding cloning steps. However, repetitive inclusion of a reporter gene is an incompressible cost and sometimes accounts for most of the synthesized DNA length. Here we present reporter systems based on split-GFP systems that reassemble into functional fluorescent proteins and can be used to monitor gene expression in E. coli TXTL. The 135 bp GFP10-11 fragment produces a fluorescent signal comparable to its full-length GFP counterpart when reassembling with its complementary protein synthesized from the 535 bp fragment expressed in TXTL. We show that split reporters can be used to characterize promoter libraries, with data qualitatively comparable to full-length GFP and matching in vivo expression measurements. We also use split reporters as small fusion tags to measure the TXTL protein and peptide production yield. Finally, we generalize our concept by providing a luminescent split reporter based on split-nanoluciferase. The ∼80% gene sequence length reduction afforded by split reporters lowers synthesis costs and liberates space for testing larger devices while producing a reliable output. In the peptide production context, the small size of split reporters compared with full-length GFP is less likely to bias peptide solubility assays. We anticipate that split reporters will facilitate rapid and cost-efficient genetic device prototyping, protein production, and interaction assays.
Collapse
Affiliation(s)
- Antoine Lévrier
- Centre de Biologie Structurale (CBS), University of Montpellier, INSERM U1054, CNRS UMR5048, Montpellier 34090, France
- School of Physics and Astronomy, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Université Paris Cité, INSERM U1284, Center for Research and Interdisciplinarity, F-75006 Paris, France
| | - Julien Capin
- Centre de Biologie Structurale (CBS), University of Montpellier, INSERM U1054, CNRS UMR5048, Montpellier 34090, France
| | - Pauline Mayonove
- Centre de Biologie Structurale (CBS), University of Montpellier, INSERM U1054, CNRS UMR5048, Montpellier 34090, France
| | - Ioannis-Ilie Karpathakis
- School of Physics and Astronomy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Peter Voyvodic
- Centre de Biologie Structurale (CBS), University of Montpellier, INSERM U1054, CNRS UMR5048, Montpellier 34090, France
| | - Angelique DeVisch
- Centre de Biologie Structurale (CBS), University of Montpellier, INSERM U1054, CNRS UMR5048, Montpellier 34090, France
| | - Ana Zuniga
- Centre de Biologie Structurale (CBS), University of Montpellier, INSERM U1054, CNRS UMR5048, Montpellier 34090, France
| | - Martin Cohen-Gonsaud
- Centre de Biologie Structurale (CBS), University of Montpellier, INSERM U1054, CNRS UMR5048, Montpellier 34090, France
| | - Stéphanie Cabantous
- Cancer Research Center of Toulouse (CRCT), Inserm, Université de Toulouse, UPS, CNRS, Toulouse 31037, France
| | - Vincent Noireaux
- School of Physics and Astronomy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jerome Bonnet
- Centre de Biologie Structurale (CBS), University of Montpellier, INSERM U1054, CNRS UMR5048, Montpellier 34090, France
| |
Collapse
|
3
|
Groseclose T, Kober EA, Clark M, Moore B, Banerjee S, Bemmer V, Beckham GT, Pickford AR, Dale TT, Nguyen HB. A High-Throughput Screening Platform for Engineering Poly(ethylene Terephthalate) Hydrolases. ACS Catal 2024; 14:14622-14638. [PMID: 39386920 PMCID: PMC11459431 DOI: 10.1021/acscatal.4c04321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/28/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024]
Abstract
The ability of enzymes to hydrolyze the ubiquitous polyester, poly(ethylene terephthalate) (PET), has enabled the potential for bioindustrial recycling of this waste plastic. To date, many of these PET hydrolases have been engineered for improved catalytic activity and stability, but current screening methods have limitations in screening large libraries, including under high-temperature conditions. Here, we developed a platform that can simultaneously interrogate PET hydrolase libraries of 104-105 variants (per round) for protein solubility, thermostability, and activity via paired, plate-based split green fluorescent protein and model substrate screens. We then applied this platform to improve the performance of a benchmark PET hydrolase, leaf-branch compost cutinase, by directed evolution. Our engineered enzyme exhibited higher catalytic activity relative to the benchmark, LCC-ICCG, on amorphous PET film coupon substrates (∼9.4% crystallinity) in pH-controlled bioreactors at both 65 °C (8.5% higher conversion at 48 h and 38% higher maximum rate, at 2.9% substrate loading) and 68 °C (11.2% higher conversion at 48 h and 43% higher maximum rate, at 16.5% substrate loading), up to 48 h, highlighting the potential of this screening platform to accelerate enzyme development for PET recycling.
Collapse
Affiliation(s)
- Thomas
M. Groseclose
- Bioscience
Division, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
- BOTTLE
Consortium, Golden, Colorado 80401, United States
| | - Erin A. Kober
- Bioscience
Division, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
- BOTTLE
Consortium, Golden, Colorado 80401, United States
| | - Matilda Clark
- BOTTLE
Consortium, Golden, Colorado 80401, United States
- Centre
for Enzyme Innovation, School of the Environmental and Life Sciences, University of Portsmouth, Portsmouth, PO1 2DT, U.K.
| | - Benjamin Moore
- BOTTLE
Consortium, Golden, Colorado 80401, United States
- Centre
for Enzyme Innovation, School of the Environmental and Life Sciences, University of Portsmouth, Portsmouth, PO1 2DT, U.K.
| | - Shounak Banerjee
- Bioscience
Division, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
- BOTTLE
Consortium, Golden, Colorado 80401, United States
| | - Victoria Bemmer
- BOTTLE
Consortium, Golden, Colorado 80401, United States
- Centre
for Enzyme Innovation, School of the Environmental and Life Sciences, University of Portsmouth, Portsmouth, PO1 2DT, U.K.
| | - Gregg T. Beckham
- BOTTLE
Consortium, Golden, Colorado 80401, United States
- Renewable
Resources and Enabling Sciences Center, National Renewable Energy Laboratory, Golden, Colorado 80401, United States
| | - Andrew R. Pickford
- BOTTLE
Consortium, Golden, Colorado 80401, United States
- Centre
for Enzyme Innovation, School of the Environmental and Life Sciences, University of Portsmouth, Portsmouth, PO1 2DT, U.K.
| | - Taraka T. Dale
- Bioscience
Division, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
- BOTTLE
Consortium, Golden, Colorado 80401, United States
| | - Hau B. Nguyen
- Bioscience
Division, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
- BOTTLE
Consortium, Golden, Colorado 80401, United States
| |
Collapse
|
4
|
Hung L, Terwilliger TC, Waldo GS, Nguyen HB. Engineering highly stable variants of Corynactis californica green fluorescent proteins. Protein Sci 2024; 33:e4886. [PMID: 38151801 PMCID: PMC10804665 DOI: 10.1002/pro.4886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/30/2023] [Accepted: 12/21/2023] [Indexed: 12/29/2023]
Abstract
Fluorescent proteins (FPs) are versatile biomarkers that facilitate effective detection and tracking of macromolecules of interest in real time. Engineered FPs such as superfolder green fluorescent protein (sfGFP) and superfolder Cherry (sfCherry) have exceptional refolding capability capable of delivering fluorescent readout in harsh environments where most proteins lose their native functions. Our recent work on the development of a split FP from a species of strawberry anemone, Corynactis californica, delivered pairs of fragments with up to threefold faster complementation than split GFP. We present the biophysical, biochemical, and structural characteristics of five full-length variants derived from these split C. californica GFP (ccGFP). These ccGFP variants are more tolerant under chemical denaturation with up to 8 kcal/mol lower unfolding free energy than that of the sfGFP. It is likely that some of these ccGFP variants could be suitable as biomarkers under more adverse environments where sfGFP fails to survive. A structural analysis suggests explanations of the variations in stabilities among the ccGFP variants.
Collapse
Affiliation(s)
- Li‐Wei Hung
- Bioscience DivisionMS M888, Los Alamos National LaboratoryLos AlamosNew MexicoUSA
| | - Thomas C. Terwilliger
- Bioscience DivisionMS M888, Los Alamos National LaboratoryLos AlamosNew MexicoUSA
- New Mexico ConsortiumLos AlamosNew MexicoUSA
| | - Geoffrey S. Waldo
- Bioscience DivisionMS M888, Los Alamos National LaboratoryLos AlamosNew MexicoUSA
| | - Hau B. Nguyen
- Bioscience DivisionMS M888, Los Alamos National LaboratoryLos AlamosNew MexicoUSA
| |
Collapse
|
5
|
Kono Y, Adam SA, Sato Y, Reddy KL, Zheng Y, Medalia O, Goldman RD, Kimura H, Shimi T. Nucleoplasmic lamin C rapidly accumulates at sites of nuclear envelope rupture with BAF and cGAS. J Cell Biol 2022; 221:e202201024. [PMID: 36301259 PMCID: PMC9617480 DOI: 10.1083/jcb.202201024] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 07/14/2022] [Accepted: 09/06/2022] [Indexed: 12/13/2022] Open
Abstract
In mammalian cell nuclei, the nuclear lamina (NL) underlies the nuclear envelope (NE) to maintain nuclear structure. The nuclear lamins, the major structural components of the NL, are involved in the protection against NE rupture induced by mechanical stress. However, the specific role of the lamins in repair of NE ruptures has not been fully determined. Our analyses using immunofluorescence and live-cell imaging revealed that the nucleoplasmic pool of lamin C rapidly accumulated at sites of NE rupture induced by laser microirradiation in mouse embryonic fibroblasts. The accumulation of lamin C at the rupture sites required both the immunoglobulin-like fold domain that binds to barrier-to-autointegration factor (BAF) and a nuclear localization signal. The accumulation of nuclear BAF and cytoplasmic cyclic GMP-AMP synthase (cGAS) at the rupture sites was in part dependent on lamin A/C. These results suggest that nucleoplasmic lamin C, BAF, and cGAS concertedly accumulate at sites of NE rupture for rapid repair.
Collapse
Affiliation(s)
- Yohei Kono
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Stephen A. Adam
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Yuko Sato
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Karen L. Reddy
- Department of Biological Chemistry, Johns Hopkins University, Baltimore, MD
| | - Yixian Zheng
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Robert D. Goldman
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Hiroshi Kimura
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
- World Research Hub Initiative, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Takeshi Shimi
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
- World Research Hub Initiative, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
6
|
Zinzula L, Mereu AM, Orsini M, Seeleitner C, Bracher A, Nagy I, Baumeister W. Ebola and Marburg virus VP35 coiled-coil validated as antiviral target by tripartite split-GFP complementation. iScience 2022; 25:105354. [PMID: 36325051 PMCID: PMC9619376 DOI: 10.1016/j.isci.2022.105354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/29/2022] [Accepted: 10/11/2022] [Indexed: 11/30/2022] Open
Abstract
Ebola virus (EBOV) and Marburg virus (MARV) are highly pathogenic viruses in humans, against which approved antivirals are lacking. During EBOV and MARV infection, coiled-coil mediated oligomerization is essential for the virion protein 35 (VP35) polymerase co-factor function and type I interferon antagonism, making VP35 coiled-coil an elective drug target. We established a tripartite split-green fluorescent protein (GFP) fluorescence complementation (FC) system based on recombinant GFP-tagged EBOV and MARV VP35, which probes VP35 coiled-coil assembly by monitoring fluorescence on E. coli colonies, or in vitro in 96/384-multiwell. Oligomerization-defective VP35 mutants showed that correct coiled-coil knobs-into-holes pairing within VP35 oligomer is pre-requisite for GFP tags and GFP detector to reconstitute fluorescing full-length GFP. The method was validated by screening a small compound library, which identified Myricetin and 4,5,6,7-Tetrabromobenzotriazole as inhibitors of EBOV and MARV VP35 oligomerization-dependent FC with low-micromolar IC50 values. These findings substantiate the VP35 coiled-coil value as antiviral target. Ebola and Marburg virus VP35 oligomerize via trimeric and tetrameric coiled-coil VP35 coiled-coil assembly triggers fluorescence of a tripartite split-GFP system Mutations perturbing VP35 coiled-coil hamper split-GFP complementation Myricetin and TBBT inhibit split-GFP complementation mediated by VP35 coiled-coil
Collapse
Affiliation(s)
- Luca Zinzula
- The Max-Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany
- Corresponding author
| | - Angela Maria Mereu
- The Max-Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Massimiliano Orsini
- Istituto Zooprofilattico Sperimentale delle Venezie, Department of Risk Analysis and Public Health Surveillance, Viale dell’Università 10, 35020 Legnaro, Italy
| | - Christine Seeleitner
- The Max-Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Andreas Bracher
- The Max-Planck Institute of Biochemistry, Department of Cellular Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - István Nagy
- The Max-Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Wolfgang Baumeister
- The Max-Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany
- Corresponding author
| |
Collapse
|
7
|
Bruguera ES, Mahoney JP, Weis WI. Reconstitution of purified membrane protein dimers in lipid nanodiscs with defined stoichiometry and orientation using a split GFP tether. J Biol Chem 2022; 298:101628. [PMID: 35074428 PMCID: PMC8980801 DOI: 10.1016/j.jbc.2022.101628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Many membrane proteins function as dimers or larger oligomers, including transporters, channels, certain signaling receptors, and adhesion molecules. In some cases, the interactions between individual proteins may be weak and/or dependent on specific lipids, such that detergent solubilization used for biochemical and structural studies disrupts functional oligomerization. Solubilized membrane protein oligomers can be captured in lipid nanodiscs, but this is an inefficient process that can produce stoichiometrically and topologically heterogeneous preparations. Here, we describe a technique to obtain purified homogeneous membrane protein dimers in nanodiscs using a split GFP (sGFP) tether. Complementary sGFP tags associate to tether the coexpressed dimers and control both stoichiometry and orientation within the nanodiscs, as assessed by quantitative Western blotting and negative-stain EM. The sGFP tether confers several advantages over other methods: it is highly stable in solution and in SDS-PAGE, which facilitates screening of dimer expression and purification by fluorescence, and also provides a dimer-specific purification handle for use with GFP nanobody–conjugated resin. We used this method to purify a Frizzled-4 homodimer and a Frizzled-4/low-density lipoprotein receptor–related protein 6 heterodimer in nanodiscs. These examples demonstrate the utility and flexibility of this method, which enables subsequent mechanistic molecular and structural studies of membrane protein pairs.
Collapse
Affiliation(s)
- Elise S Bruguera
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine; Stanford, CA 94305
| | - Jacob P Mahoney
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine; Stanford, CA 94305
| | - William I Weis
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine; Stanford, CA 94305.
| |
Collapse
|
8
|
Engineering an efficient and bright split Corynactis californica green fluorescent protein. Sci Rep 2021; 11:18440. [PMID: 34531533 PMCID: PMC8445986 DOI: 10.1038/s41598-021-98149-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/03/2021] [Indexed: 11/08/2022] Open
Abstract
Split green fluorescent protein (GFP) has been used in a panoply of cellular biology applications to study protein translocation, monitor protein solubility and aggregation, detect protein–protein interactions, enhance protein crystallization, and even map neuron contacts. Recent work shows the utility of split fluorescent proteins for large scale labeling of proteins in cells using CRISPR, but sets of efficient split fluorescent proteins that do not cross-react are needed for multiplexing experiments. We present a new monomeric split green fluorescent protein (ccGFP) engineered from a tetrameric GFP found in Corynactis californica, a bright red colonial anthozoan similar to sea anemones and scleractinian stony corals. Split ccGFP from C. californica complements up to threefold faster compared to the original Aequorea victoria split GFP and enable multiplexed labeling with existing A. victoria split YFP and CFP.
Collapse
|
9
|
Current pivotal strategies leading a difficult target protein to a sample suitable for crystallographic analysis. Biochem Soc Trans 2020; 48:1661-1673. [PMID: 32677661 DOI: 10.1042/bst20200106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/15/2022]
Abstract
Crystallographic structural analysis is an essential method for the determination of protein structure. However, crystallization of a protein of interest is the most difficult process in the analysis. The process is often hampered during the sample preparation, including expression and purification. Even after a sample has been purified, not all candidate proteins crystallize. In this mini-review, the current methodologies used to overcome obstacles encountered during protein crystallization are sorted. Specifically, the strategy for an effective crystallization is compared with a pipeline where various expression hosts and constructs, purification and crystallization conditions, and crystallization chaperones as target-specific binder proteins are assessed by a precrystallization screening. These methodologies are also developed continuously to improve the process. The described methods are useful for sample preparation in crystallographic analysis and other structure determination techniques, such as cryo-electron microscopy.
Collapse
|
10
|
Custodio R, Johnson E, Liu G, Tang CM, Exley RM. Commensal Neisseria cinerea impairs Neisseria meningitidis microcolony development and reduces pathogen colonisation of epithelial cells. PLoS Pathog 2020; 16:e1008372. [PMID: 32208456 PMCID: PMC7092958 DOI: 10.1371/journal.ppat.1008372] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/31/2020] [Indexed: 12/16/2022] Open
Abstract
It is increasingly being recognised that the interplay between commensal and pathogenic bacteria can dictate the outcome of infection. Consequently, there is a need to understand how commensals interact with their human host and influence pathogen behaviour at epithelial surfaces. Neisseria meningitidis, a leading cause of sepsis and meningitis, exclusively colonises the human nasopharynx and shares this niche with several other Neisseria species, including the commensal Neisseria cinerea. Here, we demonstrate that during adhesion to human epithelial cells N. cinerea co-localises with molecules that are also recruited by the meningococcus, and show that, similar to N. meningitidis, N. cinerea forms dynamic microcolonies on the cell surface in a Type four pilus (Tfp) dependent manner. Finally, we demonstrate that N. cinerea colocalises with N. meningitidis on the epithelial cell surface, limits the size and motility of meningococcal microcolonies, and impairs the effective colonisation of epithelial cells by the pathogen. Our data establish that commensal Neisseria can mimic and affect the behaviour of a pathogen on epithelial cell surfaces.
Collapse
Affiliation(s)
- Rafael Custodio
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Errin Johnson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Guangyu Liu
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Christoph M. Tang
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Rachel M. Exley
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
11
|
Porous crystals as scaffolds for structural biology. Curr Opin Struct Biol 2020; 60:85-92. [PMID: 31896427 DOI: 10.1016/j.sbi.2019.12.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/15/2019] [Accepted: 12/05/2019] [Indexed: 12/22/2022]
Abstract
Molecular scaffolds provide routes to otherwise inaccessible organized states of matter. Scaffolds that are crystalline can be observed in atomic detail using diffraction, along with any guest molecules that have adopted coherent structures therein. This approach, scaffold-assisted structure determination, is not yet routine. However, with varying degrees of guest immobilization, porous crystal scaffolds have recently been decorated with guest molecules. Herein we analyze recent milestones, compare the relative advantages and challenges of different types of scaffold crystals, and weigh the merits of diverse guest installation strategies.
Collapse
|
12
|
Feng S, Varshney A, Coto Villa D, Modavi C, Kohler J, Farah F, Zhou S, Ali N, Müller JD, Van Hoven MK, Huang B. Bright split red fluorescent proteins for the visualization of endogenous proteins and synapses. Commun Biol 2019; 2:344. [PMID: 31552297 PMCID: PMC6749000 DOI: 10.1038/s42003-019-0589-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 08/26/2019] [Indexed: 12/22/2022] Open
Abstract
Self-associating split fluorescent proteins (FPs) are split FPs whose two fragments spontaneously associate to form a functional FP. They have been widely used for labeling proteins, scaffolding protein assembly and detecting cell-cell contacts. Recently developments have expanded the palette of self-associating split FPs beyond the original split GFP1-10/11. However, these new ones have suffered from suboptimal fluorescence signal after complementation. Here, by investigating the complementation process, we have demonstrated two approaches to improve split FPs: assistance through SpyTag/SpyCatcher interaction and directed evolution. The latter has yielded two split sfCherry3 variants with substantially enhanced overall brightness, facilitating the tagging of endogenous proteins by gene editing. Based on sfCherry3, we have further developed a new red-colored trans-synaptic marker called Neuroligin-1 sfCherry3 Linker Across Synaptic Partners (NLG-1 CLASP) for multiplexed visualization of neuronal synapses in living C. elegans, demonstrating its broad applications.
Collapse
Affiliation(s)
- Siyu Feng
- The UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA 94143 USA
| | - Aruna Varshney
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192 USA
| | - Doris Coto Villa
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192 USA
| | - Cyrus Modavi
- Department of Bioengineering and Therapeutic Sciences, University of California in San Francisco, San Francisco, CA 94143 USA
| | - John Kohler
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455 USA
| | - Fatima Farah
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192 USA
| | - Shuqin Zhou
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084 China
- Department of Pharmaceutical Chemistry, University of California in San Francisco, San Francisco, CA 94143 USA
| | - Nebat Ali
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192 USA
| | - Joachim D. Müller
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455 USA
| | - Miri K. Van Hoven
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192 USA
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California in San Francisco, San Francisco, CA 94143 USA
- Department Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143 USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158 USA
| |
Collapse
|
13
|
Pedelacq JD, Cabantous S. Development and Applications of Superfolder and Split Fluorescent Protein Detection Systems in Biology. Int J Mol Sci 2019; 20:ijms20143479. [PMID: 31311175 PMCID: PMC6678664 DOI: 10.3390/ijms20143479] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/08/2019] [Accepted: 07/08/2019] [Indexed: 01/08/2023] Open
Abstract
Molecular engineering of the green fluorescent protein (GFP) into a robust and stable variant named Superfolder GFP (sfGFP) has revolutionized the field of biosensor development and the use of fluorescent markers in diverse area of biology. sfGFP-based self-associating bipartite split-FP systems have been widely exploited to monitor soluble expression in vitro, localization, and trafficking of proteins in cellulo. A more recent class of split-FP variants, named « tripartite » split-FP, that rely on the self-assembly of three GFP fragments, is particularly well suited for the detection of protein–protein interactions. In this review, we describe the different steps and evolutions that have led to the diversification of superfolder and split-FP reporter systems, and we report an update of their applications in various areas of biology, from structural biology to cell biology.
Collapse
Affiliation(s)
- Jean-Denis Pedelacq
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France.
| | - Stéphanie Cabantous
- Centre de Recherche en Cancérologie de Toulouse (CRCT), Inserm, Université Paul Sabatier-Toulouse III, CNRS, 31037 Toulouse, France.
| |
Collapse
|
14
|
Ferreira AV, Antunes E, Ribeiro A, Matamá T, Azoia NG, Cunha J, Cavaco-Paulo A. Design of a chromogenic substrate for elastase based on split GFP system-Proof of concept for colour switch sensors. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2019; 22:e00324. [PMID: 31049301 PMCID: PMC6479270 DOI: 10.1016/j.btre.2019.e00324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/15/2019] [Accepted: 03/15/2019] [Indexed: 06/09/2023]
Abstract
Recent studies have demonstrated that human neutrophil elastase (HNE) can be used as marker for inflammation/infection of chronic wounds since it was found to be present in high concentration in exudate collected from chronic wounds. Biosensors used in wound care benefit from a chromogenic signalling due to the readiness of signal interpretation, but the most common use faint yellow chromogenic molecules such as p-nitroaniline (pNa). In addition, if to be converted into smart dressings, the colour of the detection system should not be masked by the exudate's colour. In this work, we designed a chromogenic substrate for HNE aiming to be incorporated in a smart dressing as a colour switch sensor. The substrate was developed using the GFP-like chromoprotein ultramarine (UM), following the split GFP technology. The cleavage sequence for HNE (Ala-Ala-Pro-Val) was embedded into the sensing moiety of the substrate corresponding to the 11th β-sheet. In the presence of HNE, the 11th β-sheet is able to interact to the signalling moiety composed of the β1-β10 incomplete barrel, allowing the re-establishment of the chromophore environment and, hence, the colour production. Structural homology and molecular dynamics simulations were conducted to aid on the disclosure of the structural changes that are the base of the mechanism of action of this HNE switch substrate. Our findings explore the possible application of GFP-like chromogenic sensors in point-of-care devices for the evaluation of the wounds status, representing a major step in the medical field.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Artur Cavaco-Paulo
- Centre of Biological Engineering (CEB), University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal
| |
Collapse
|
15
|
Detecting In-Situ oligomerization of engineered STIM1 proteins by diffraction-limited optical imaging. PLoS One 2019; 14:e0213655. [PMID: 30908505 PMCID: PMC6433367 DOI: 10.1371/journal.pone.0213655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 02/26/2019] [Indexed: 11/26/2022] Open
Abstract
Several signaling proteins require self-association of individual monomer units to be activated for triggering downstream signaling cascades in cells. Methods that allow visualizing their underlying molecular mechanisms will immensely benefit cell biology. Using enhanced Green Fluorescent Protein (eGFP) complementation, here I present a functional imaging approach for visualizing the protein-protein interaction in cells. Activation mechanism of an ER (endoplasmic reticulum) resident Ca2+ sensor, STIM1 (Stromal Interaction Molecule 1) that regulates store-operated Ca2+ entry in cells is considered as a model system. Co-expression of engineered full-length human STIM1 (ehSTIM1) with N-terminal complementary split eGFP pairs in mammalian cells fluoresces to form ‘puncta’ upon a drop in ER lumen Ca2+ concentration. Quantization of discrete fluorescent intensities of ehSTIM1 molecules at a diffraction-limited resolution revealed a diverse set of intensity levels not exceeding six-fold. Detailed screening of the ehSTIM1 molecular entities characterized by one to six fluorescent emitters across various in-plane sections shows a greater probability of occurrence for entities with six emitters in the vicinity of the plasma membrane (PM) than at the interior sections. However, the number density of entities with six emitters was lesser than that of others localized close to the PM. This finding led to hypothesize that activated ehSTIM1 dimers perhaps oligomerize in bundles ranging from 1–6 with an increased propensity for the occurrence of hexamers of ehSTIM1 dimer units close to PM even when its partner protein, ORAI1 (PM resident Ca2+ channel) is not sufficiently over-expressed in cells. The experimental data presented here provide direct evidence for luminal domain association of ehSTIM1 monomer units to trigger activation and allow enumerating various oligomers of ehSTIM1 in cells.
Collapse
|
16
|
Zhang Q, Schepis A, Huang H, Yang J, Ma W, Torra J, Zhang SQ, Yang L, Wu H, Nonell S, Dong Z, Kornberg TB, Coughlin SR, Shu X. Designing a Green Fluorogenic Protease Reporter by Flipping a Beta Strand of GFP for Imaging Apoptosis in Animals. J Am Chem Soc 2019; 141:4526-4530. [PMID: 30821975 PMCID: PMC6486793 DOI: 10.1021/jacs.8b13042] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A family of proteases called caspases mediate apoptosis signaling in animals. We report a GFP-based fluorogenic protease reporter, dubbed "FlipGFP", by flipping a beta strand of the GFP. Upon protease activation and cleavage, the beta strand is restored, leading to reconstitution of the GFP and fluorescence. FlipGFP-based TEV protease reporter achieves 100-fold fluorescence change. A FlipGFP-based executioner caspase reporter visualized apoptosis in live zebrafish embryos with spatiotemporal resolution. FlipGFP also visualized apoptotic cells in the midgut of Drosophila. Thus, the FlipGFP-based caspase reporter will be useful for monitoring apoptosis during animal development and for designing reporters of proteases beyond caspases. The design strategy can be further applied to a red fluorescent protein for engineering a red fluorogenic protease reporter.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, California 94158, United States
- Cardiovascular Research Institute, University of California–San Francisco, San Francisco, California 94158, United States
| | - Antonino Schepis
- Cardiovascular Research Institute, University of California–San Francisco, San Francisco, California 94158, United States
| | - Hai Huang
- Cardiovascular Research Institute, University of California–San Francisco, San Francisco, California 94158, United States
| | - Junjiao Yang
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, California 94158, United States
- Cardiovascular Research Institute, University of California–San Francisco, San Francisco, California 94158, United States
| | - Wen Ma
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
| | - Joaquim Torra
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, California 94158, United States
- Cardiovascular Research Institute, University of California–San Francisco, San Francisco, California 94158, United States
- institut Quimic de Sarria, Universitat Ramon Llull, Via Augusta 390, 08017 Barcelona, Spain
| | - Shao-Qing Zhang
- Department of Neurology, Harvard Medical School and MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, Massachusetts 02129, United States
| | - Lina Yang
- Bio-Medical Center, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Haifan Wu
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, California 94158, United States
- Cardiovascular Research Institute, University of California–San Francisco, San Francisco, California 94158, United States
| | - Santi Nonell
- institut Quimic de Sarria, Universitat Ramon Llull, Via Augusta 390, 08017 Barcelona, Spain
| | - Zhiqiang Dong
- Bio-Medical Center, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Thomas B. Kornberg
- Cardiovascular Research Institute, University of California–San Francisco, San Francisco, California 94158, United States
| | - Shaun R. Coughlin
- Cardiovascular Research Institute, University of California–San Francisco, San Francisco, California 94158, United States
| | - Xiaokun Shu
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, California 94158, United States
- Cardiovascular Research Institute, University of California–San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
17
|
Abstract
Many proteins can be split into fragments that spontaneously reassemble, without covalent linkage, into a functional protein. For split green fluorescent proteins (GFPs), fragment reassembly leads to a fluorescent readout, which has been widely used to investigate protein-protein interactions. We review the scope and limitations of this approach as well as other diverse applications of split GFPs as versatile sensors, molecular glues, optogenetic tools, and platforms for photophysical studies.
Collapse
Affiliation(s)
- Matthew G Romei
- Department of Chemistry, Stanford University, Stanford, California 94305, USA; ,
| | - Steven G Boxer
- Department of Chemistry, Stanford University, Stanford, California 94305, USA; ,
| |
Collapse
|
18
|
Koh HG, Kang NK, Kim EK, Jeon S, Shin SE, Lee B, Chang YK. Advanced multigene expression system for Nannochloropsis salina using 2A self-cleaving peptides. J Biotechnol 2018; 278:39-47. [PMID: 29715488 DOI: 10.1016/j.jbiotec.2018.04.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 11/19/2022]
Abstract
Even though there has been much interest in genetic engineering of microalgae, its progress has been slow due to the difficulty and limitation of available techniques. Currently, genetic modification in most microalgal strains is confined to single gene transformation. Here, a multigene expression system for the oleaginous model strain Nannochloropsis salina was developed with glycine-serine-glycine spacer linked 2A self-cleaving peptides (2A) for the first time. An efficiency test of the four most widely used 2As revealed that two different types of 2As T2A and E2A have the best performance in N. salina with a maximum cleavage rate of nearly 45%. The system was able to express the linked sequence of the selection marker shble and the fluorescence protein sfCherry with intact functions. Because 2A enabled multigene expression in the single cassette form, the use of 2A also reduced the vector size, which along with the stronger promoter resulted in a 9-fold increase in the transformation efficiency. Furthermore, confirmative screening accuracy of more than 90% was observed. Hence, the 2A applied vector system is expected to be beneficial in microalgal research field because it enables multigene expression as well as offering improved transformation and screening efficiency.
Collapse
Affiliation(s)
- Hyun Gi Koh
- Department of Chemical and Biomolecular Engineering, KAIST, Daejeon 34141, Republic of Korea.
| | - Nam Kyu Kang
- Advanced Biomass R&D Center (ABC), Daejeon 34141, Republic of Korea.
| | - Eun Kyung Kim
- Advanced Biomass R&D Center (ABC), Daejeon 34141, Republic of Korea.
| | - Seungjib Jeon
- Department of Chemical and Biomolecular Engineering, KAIST, Daejeon 34141, Republic of Korea.
| | - Sung-Eun Shin
- Department of Chemical and Biomolecular Engineering, KAIST, Daejeon 34141, Republic of Korea.
| | - Bongsoo Lee
- Department of Chemical and Biomolecular Engineering, KAIST, Daejeon 34141, Republic of Korea.
| | - Yong Keun Chang
- Department of Chemical and Biomolecular Engineering, KAIST, Daejeon 34141, Republic of Korea; Advanced Biomass R&D Center (ABC), Daejeon 34141, Republic of Korea.
| |
Collapse
|
19
|
Improved split fluorescent proteins for endogenous protein labeling. Nat Commun 2017; 8:370. [PMID: 28851864 PMCID: PMC5575300 DOI: 10.1038/s41467-017-00494-8] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 06/30/2017] [Indexed: 12/22/2022] Open
Abstract
Self-complementing split fluorescent proteins (FPs) have been widely used for protein labeling, visualization of subcellular protein localization, and detection of cell–cell contact. To expand this toolset, we have developed a screening strategy for the direct engineering of self-complementing split FPs. Via this strategy, we have generated a yellow–green split-mNeonGreen21–10/11 that improves the ratio of complemented signal to the background of FP1–10-expressing cells compared to the commonly used split GFP1–10/11; as well as a 10-fold brighter red-colored split-sfCherry21–10/11. Based on split sfCherry2, we have engineered a photoactivatable variant that enables single-molecule localization-based super-resolution microscopy. We have demonstrated dual-color endogenous protein tagging with sfCherry211 and GFP11, revealing that endoplasmic reticulum translocon complex Sec61B has reduced abundance in certain peripheral tubules. These new split FPs not only offer multiple colors for imaging interaction networks of endogenous proteins, but also hold the potential to provide orthogonal handles for biochemical isolation of native protein complexes. Split fluorescent proteins (FPs) have been widely used to visualise proteins in cells. Here the authors develop a screen for engineering new split FPs, and report a yellow-green split-mNeonGreen2 with reduced background, a red split-sfCherry2 for multicolour labeling, and its photoactivatable variant for super-resolution use.
Collapse
|
20
|
Knapp A, Ripphahn M, Volkenborn K, Skoczinski P, Jaeger KE. Activity-independent screening of secreted proteins using split GFP. J Biotechnol 2017; 258:110-116. [PMID: 28619616 DOI: 10.1016/j.jbiotec.2017.05.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/31/2017] [Accepted: 05/31/2017] [Indexed: 01/16/2023]
Abstract
The large-scale industrial production of proteins requires efficient secretion, as provided, for instance, by the Sec system of Gram-positive bacteria. Protein engineering approaches to optimize secretion often involve the screening of large libraries, e.g. comprising a target protein fused to many different signal peptides. Respective high-throughput screening methods are usually based on photometric or fluorimetric assays enabling fast and simple determination of enzymatic activities. Here, we report on an alternative method for quantification of secreted proteins based on the split GFP assay. We analyzed the secretion by Bacillus subtilis of a homologous lipase and a heterologous cutinase by determination of GFP fluorescence and enzyme activity assays. Furthermore, we identified from a signal peptide library a variant of the biotechnologically relevant B. subtilis protein swollenin EXLX1 with up to 5-fold increased secretion. Our results demonstrate that the split GFP assay can be used to monitor secretion of enzymatic and non-enzymatic proteins in B. subtilis in a high-throughput manner.
Collapse
Affiliation(s)
- Andreas Knapp
- Institute of Molecular Enzyme Technology, Heinrich -Heine -University Düsseldorf, Forschungszentrum Jülich, D-52426 Jülich, Germany
| | - Myriam Ripphahn
- Institute of Molecular Enzyme Technology, Heinrich -Heine -University Düsseldorf, Forschungszentrum Jülich, D-52426 Jülich, Germany
| | - Kristina Volkenborn
- Institute of Molecular Enzyme Technology, Heinrich -Heine -University Düsseldorf, Forschungszentrum Jülich, D-52426 Jülich, Germany
| | - Pia Skoczinski
- Institute of Molecular Enzyme Technology, Heinrich -Heine -University Düsseldorf, Forschungszentrum Jülich, D-52426 Jülich, Germany
| | - Karl-Erich Jaeger
- Institute of Molecular Enzyme Technology, Heinrich -Heine -University Düsseldorf, Forschungszentrum Jülich, D-52426 Jülich, Germany; Institute of Bio- and Geosciences IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, D-52428 Jülich, Germany.
| |
Collapse
|
21
|
Sjöhamn J, Båth P, Neutze R, Hedfalk K. Applying bimolecular fluorescence complementation to screen and purify aquaporin protein:protein complexes. Protein Sci 2016; 25:2196-2208. [PMID: 27643892 PMCID: PMC5119558 DOI: 10.1002/pro.3046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/14/2016] [Accepted: 09/14/2016] [Indexed: 12/22/2022]
Abstract
Protein:protein interactions play key functional roles in the molecular machinery of the cell. A major challenge for structural biology is to gain high‐resolution structural insight into how membrane protein function is regulated by protein:protein interactions. To this end we present a method to express, detect, and purify stable membrane protein complexes that are suitable for further structural characterization. Our approach utilizes bimolecular fluorescence complementation (BiFC), whereby each protein of an interaction pair is fused to nonfluorescent fragments of yellow fluorescent protein (YFP) that combine and mature as the complex is formed. YFP thus facilitates the visualization of protein:protein interactions in vivo, stabilizes the assembled complex, and provides a fluorescent marker during purification. This technique is validated by observing the formation of stable homotetramers of human aquaporin 0 (AQP0). The method's broader applicability is demonstrated by visualizing the interactions of AQP0 and human aquaporin 1 (AQP1) with the cytoplasmic regulatory protein calmodulin (CaM). The dependence of the AQP0‐CaM complex on the AQP0 C‐terminus is also demonstrated since the C‐terminal truncated construct provides a negative control. This screening approach may therefore facilitate the production and purification of membrane protein:protein complexes for later structural studies by X‐ray crystallography or single particle electron microscopy.
Collapse
Affiliation(s)
- Jennie Sjöhamn
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, SE-405 30, Sweden
| | - Petra Båth
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, SE-405 30, Sweden
| | - Richard Neutze
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, SE-405 30, Sweden
| | - Kristina Hedfalk
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, SE-405 30, Sweden
| |
Collapse
|
22
|
Kamiyama D, Sekine S, Barsi-Rhyne B, Hu J, Chen B, Gilbert LA, Ishikawa H, Leonetti MD, Marshall WF, Weissman JS, Huang B. Versatile protein tagging in cells with split fluorescent protein. Nat Commun 2016; 7:11046. [PMID: 26988139 PMCID: PMC4802074 DOI: 10.1038/ncomms11046] [Citation(s) in RCA: 298] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 02/15/2016] [Indexed: 12/26/2022] Open
Abstract
In addition to the popular method of fluorescent protein fusion, live cell protein imaging has now seen more and more application of epitope tags. The small size of these tags may reduce functional perturbation and enable signal amplification. To address their background issue, we adapt self-complementing split fluorescent proteins as epitope tags for live cell protein labelling. The two tags, GFP11 and sfCherry11 are derived from the eleventh β-strand of super-folder GFP and sfCherry, respectively. The small size of FP11-tags enables a cost-effective and scalable way to insert them into endogenous genomic loci via CRISPR-mediated homology-directed repair. Tandem arrangement FP11-tags allows proportional enhancement of fluorescence signal in tracking intraflagellar transport particles, or reduction of photobleaching for live microtubule imaging. Finally, we show the utility of tandem GFP11-tag in scaffolding protein oligomerization. These experiments illustrate the versatility of FP11-tag as a labelling tool as well as a multimerization-control tool for both imaging and non-imaging applications. Tagging proteins with fluorescent proteins is a powerful method for both imaging and non-imaging applications. Here the authors use the eleventh β-strand of sfGFP and sfCherry as epitope tags for multicolour imaging and amplified signals by tandem arrangement; shortness of the tag enabled introduction into genomic loci using CRISPR/Cas9.
Collapse
Affiliation(s)
- Daichi Kamiyama
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, USA
| | - Sayaka Sekine
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, USA
| | - Benjamin Barsi-Rhyne
- Tetrad Graduate Program, University of California, San Francisco, California 94143, USA
| | - Jeffrey Hu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, USA
| | - Baohui Chen
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, USA
| | - Luke A Gilbert
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California 94143, USA
| | - Hiroaki Ishikawa
- Department of Biochemistry and Biophysics, University of California, San Francisco, California 94143, USA
| | - Manuel D Leonetti
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California 94143, USA
| | - Wallace F Marshall
- Department of Biochemistry and Biophysics, University of California, San Francisco, California 94143, USA
| | - Jonathan S Weissman
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California 94143, USA.,Howard Hughes Medical Institute, San Francisco, California 94143, USA
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, USA.,Department of Biochemistry and Biophysics, University of California, San Francisco, California 94143, USA
| |
Collapse
|
23
|
Heterologous overexpression of sfCherry fluorescent protein in Nannochloropsis salina. ACTA ACUST UNITED AC 2015; 8:10-15. [PMID: 28352568 PMCID: PMC4980701 DOI: 10.1016/j.btre.2015.08.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 12/22/2022]
Abstract
Heterologous sfCherry protein was expressed in N. salina for the first time. N. salina was transformed by particle bombardment. Integration site of the transgene on the genome was determined by RESDA PCR. Expression of sfCherry was confirmed by a western blotting and confocal microscopy.
Oleaginous microalgae of the Nannochloropsis genus are considered excellent candidates for biofuels and value-added products owing to their high biomass productivity and lipid content. Here, we report the first overexpression and detection of a heterologous sfCherry fluorescent protein in Nannochloropsis salina in order to develop a transformation toolbox for future genetic improvements. Particle bombardment was employed for transformation, and expression of Shble under the control of TUB and UEP promoters, cloned from N. salina, was used to confer resistance to Zeocin antibiotics, resulting in 5.9 and 4.7 transformants per 108 cells, respectively. Stable integration of the markers into the genome was confirmed using a restriction enzyme site-directed amplification (RESDA) PCR. The expression of sfCherry fluorescent protein was confirmed by Western blot analysis and confocal microscopy. These results suggest new possibilities of efficient genetic engineering of Nannochloropsis for the production of biofuels and other biochemicals.
Collapse
|
24
|
Leibly DJ, Arbing MA, Pashkov I, DeVore N, Waldo GS, Terwilliger TC, Yeates TO. A Suite of Engineered GFP Molecules for Oligomeric Scaffolding. Structure 2015; 23:1754-1768. [PMID: 26278175 DOI: 10.1016/j.str.2015.07.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 06/08/2015] [Accepted: 07/07/2015] [Indexed: 10/23/2022]
Abstract
Applications ranging from synthetic biology to protein crystallization could be advanced by facile systems for connecting multiple proteins together in predefined spatial relationships. One approach to this goal is to engineer many distinct assembly forms of a single carrier protein or scaffold, to which other proteins of interest can then be readily attached. In this work we chose GFP as a scaffold and engineered many alternative oligomeric forms, driven by either specific disulfide bond formation or metal ion addition. We generated a wide range of spatial arrangements of GFP subunits from 11 different oligomeric variants, and determined their X-ray structures in a total of 33 distinct crystal forms. Some of the oligomeric GFP variants show geometric polymorphism depending on conditions, while others show considerable geometric rigidity. Potential future applications of this system are discussed.
Collapse
Affiliation(s)
- David J Leibly
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA; UCLA-DOE Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095, USA
| | - Mark A Arbing
- UCLA-DOE Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095, USA
| | - Inna Pashkov
- UCLA-DOE Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095, USA
| | - Natasha DeVore
- Bioscience Division, Los Alamos National Laboratory, MS M888, Los Alamos, NM 87545, USA
| | - Geoffrey S Waldo
- Bioscience Division, Los Alamos National Laboratory, MS M888, Los Alamos, NM 87545, USA
| | - Thomas C Terwilliger
- Bioscience Division, Los Alamos National Laboratory, MS M888, Los Alamos, NM 87545, USA
| | - Todd O Yeates
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA; UCLA-DOE Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
25
|
Kobe B, Ve T, Williams SJ. Fusion-protein-assisted protein crystallization. Acta Crystallogr F Struct Biol Commun 2015; 71:861-9. [PMID: 26144231 PMCID: PMC4498707 DOI: 10.1107/s2053230x15011061] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 06/07/2015] [Indexed: 01/29/2023] Open
Abstract
Fusion proteins can be used directly in protein crystallization to assist crystallization in at least two different ways. In one approach, the `heterologous fusion-protein approach', the fusion partner can provide additional surface area to promote crystal contact formation. In another approach, the `fusion of interacting proteins approach', protein assemblies can be stabilized by covalently linking the interacting partners. The linker connecting the proteins plays different roles in the two applications: in the first approach a rigid linker is required to reduce conformational heterogeneity; in the second, conversely, a flexible linker is required that allows the native interaction between the fused proteins. The two approaches can also be combined. The recent applications of fusion-protein technology in protein crystallization from the work of our own and other laboratories are briefly reviewed.
Collapse
Affiliation(s)
- Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Thomas Ve
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Simon J. Williams
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland 4072, Australia
- School of Biological Sciences, Flinders University, Adelaide, South Australia 5001, Australia
| |
Collapse
|