1
|
Cai P, Casas CJ, Quintero Plancarte G, Mikawa T, Hua LL. Ipsilateral restriction of chromosome movement along a centrosome, and apical-basal axis during the cell cycle. Chromosome Res 2025; 33:1. [PMID: 39751905 PMCID: PMC11698895 DOI: 10.1007/s10577-024-09760-0] [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: 11/05/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025]
Abstract
Little is known about how distance between homologous chromosomes are controlled during the cell cycle. Here, we show that the distribution of centromere components display two discrete clusters placed to either side of the centrosome and apical/basal axis from prophase to G1 interphase. 4-Dimensional live cell imaging analysis of centromere and centrosome tracking reveals that centromeres oscillate largely within one cluster, but do not cross over to the other cluster. We propose a model of an axis-dependent ipsilateral restriction of chromosome oscillations throughout mitosis.
Collapse
Affiliation(s)
- Pingping Cai
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Christian J Casas
- Department of Biology, Sonoma State University, Rohnert Park, CA, USA
| | | | - Takashi Mikawa
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
| | - Lisa L Hua
- Department of Biology, Sonoma State University, Rohnert Park, CA, USA.
| |
Collapse
|
2
|
Grätz L, Sajkowska-Kozielewicz JJ, Wesslowski J, Kinsolving J, Bridge LJ, Petzold K, Davidson G, Schulte G, Kozielewicz P. NanoBiT- and NanoBiT/BRET-based assays allow the analysis of binding kinetics of Wnt-3a to endogenous Frizzled 7 in a colorectal cancer model. Br J Pharmacol 2024; 181:3819-3835. [PMID: 37055379 DOI: 10.1111/bph.16090] [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: 11/15/2022] [Revised: 03/06/2023] [Accepted: 04/05/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND AND PURPOSE Wnt binding to Frizzleds (FZD) is a crucial step that leads to the initiation of signalling cascades governing multiple processes during embryonic development, stem cell regulation and adult tissue homeostasis. Recent efforts have enabled us to shed light on Wnt-FZD pharmacology using overexpressed HEK293 cells. However, assessing ligand binding at endogenous receptor expression levels is important due to differential binding behaviour in a native environment. Here, we study FZD paralogue, FZD7, and analyse its interactions with Wnt-3a in live CRISPR-Cas9-edited SW480 cells typifying colorectal cancer. EXPERIMENTAL APPROACH SW480 cells were CRISPR-Cas9-edited to insert a HiBiT tag on the N-terminus of FZD7, preserving the native signal peptide. These cells were used to study eGFP-Wnt-3a association with endogenous and overexpressed HiBiT-FZD7 using NanoBiT/bioluminescence resonance energy transfer (BRET) and NanoBiT to measure ligand binding and receptor internalization. KEY RESULTS With this new assay the binding of eGFP-Wnt-3a to endogenous HiBiT-FZD7 was compared with overexpressed receptors. Receptor overexpression results in increased membrane dynamics, leading to an apparent decrease in binding on-rate and consequently in higher, up to 10 times, calculated Kd. Thus, measurements of binding affinities to FZD7 obtained in overexpressed cells are suboptimal compared with the measurements from endogenously expressing cells. CONCLUSIONS AND IMPLICATIONS Binding affinity measurements in the overexpressing cells fail to replicate ligand binding affinities assessed in a (patho)physiologically relevant context where receptor expression is lower. Therefore, future studies on Wnt-FZD7 binding should be performed using receptors expressed under endogenous promotion.
Collapse
Affiliation(s)
- Lukas Grätz
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Joanna J Sajkowska-Kozielewicz
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Janine Wesslowski
- Institute of Biological and Chemical Systems-Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Julia Kinsolving
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Lloyd J Bridge
- Department of Computer Science and Creative Technologies, University of the West England, Bristol, UK
| | - Katja Petzold
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Gary Davidson
- Institute of Biological and Chemical Systems-Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Gunnar Schulte
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Paweł Kozielewicz
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
3
|
Claff T, Mahardhika AB, Vaaßen VJ, Schlegel J, Vielmuth C, Weiße RH, Sträter N, Müller CE. Structural Insights into Partial Activation of the Prototypic G Protein-Coupled Adenosine A 2A Receptor. ACS Pharmacol Transl Sci 2024; 7:1415-1425. [PMID: 38751633 PMCID: PMC11091970 DOI: 10.1021/acsptsci.4c00051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 05/18/2024]
Abstract
The adenosine A2A receptor (A2AAR) belongs to the rhodopsin-like G protein-coupled receptor (GPCR) family, which constitutes the largest class of GPCRs. Partial agonists show reduced efficacy as compared to physiological agonists and can even act as antagonists in the presence of a full agonist. Here, we determined an X-ray crystal structure of the partial A2AAR agonist 2-amino-6-[(1H-imidazol-2-ylmethyl)sulfanyl]-4-p-hydroxyphenyl-3,5-pyridinedicarbonitrile (LUF5834) in complex with the A2AAR construct A2A-PSB2-bRIL, stabilized in its inactive conformation and being devoid of any mutations in the ligand binding pocket. The determined high-resolution structure (2.43 Å) resolved water networks and crucial binding pocket interactions. A direct hydrogen bond of the p-hydroxy group of LUF5834 with T883.36 was observed, an amino acid that was mutated to alanine in the most frequently used A2AAR crystallization constructs thus preventing the discovery of its interactions in most of the previous A2AAR co-crystal structures. G protein dissociation studies confirmed partial agonistic activity of LUF5834 as compared to that of the full agonist N-ethylcarboxamidoadenosine (NECA). In contrast to NECA, the partial agonist was still able to bind to the receptor construct locked in its inactive conformation by an S913.39K mutation, although with an affinity lower than that at the native receptor. This could explain the compound's partial agonistic activity: while full A2AAR agonists bind exclusively to the active conformation, likely following conformational selection, partial agonists bind to active as well as inactive conformations, showing higher affinity for the active conformation. This might be a general mechanism of partial agonism also applicable to other GPCRs.
Collapse
Affiliation(s)
- Tobias Claff
- PharmaCenter
Bonn & Pharmaceutical Institute, Department of Pharmaceutical
& Medicinal Chemistry, University of
Bonn, Bonn 53113, Germany
| | - Andhika B. Mahardhika
- PharmaCenter
Bonn & Pharmaceutical Institute, Department of Pharmaceutical
& Medicinal Chemistry, University of
Bonn, Bonn 53113, Germany
- Research
Training Group 2873, University of Bonn, Bonn 53121, Germany
| | - Victoria J. Vaaßen
- PharmaCenter
Bonn & Pharmaceutical Institute, Department of Pharmaceutical
& Medicinal Chemistry, University of
Bonn, Bonn 53113, Germany
| | - Jonathan
G. Schlegel
- PharmaCenter
Bonn & Pharmaceutical Institute, Department of Pharmaceutical
& Medicinal Chemistry, University of
Bonn, Bonn 53113, Germany
| | - Christin Vielmuth
- PharmaCenter
Bonn & Pharmaceutical Institute, Department of Pharmaceutical
& Medicinal Chemistry, University of
Bonn, Bonn 53113, Germany
| | - Renato H. Weiße
- Institute
of Bioanalytical Chemistry, Center for Biotechnology and Biomedicine, Leipzig University, Leipzig 04103, Germany
| | - Norbert Sträter
- Institute
of Bioanalytical Chemistry, Center for Biotechnology and Biomedicine, Leipzig University, Leipzig 04103, Germany
| | - Christa E. Müller
- PharmaCenter
Bonn & Pharmaceutical Institute, Department of Pharmaceutical
& Medicinal Chemistry, University of
Bonn, Bonn 53113, Germany
- Research
Training Group 2873, University of Bonn, Bonn 53121, Germany
| |
Collapse
|
4
|
Cai P, Casas CJ, Plancarte GQ, Mikawa T, Hua LL. Ipsilateral restriction of chromosome movement along a centrosome, and apical-basal axis during the cell cycle. RESEARCH SQUARE 2024:rs.3.rs-4283973. [PMID: 38746098 PMCID: PMC11092853 DOI: 10.21203/rs.3.rs-4283973/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Little is known about how distance between homologous chromosomes are controlled during the cell cycle. Here, we show that the distribution of centromere components display two discrete clusters placed to either side of the centrosome and apical/basal axis from prophase to G1 interphase. 4-Dimensional live cell imaging analysis of centromere and centrosome tracking reveals that centromeres oscillate largely within one cluster, but do not cross over to the other cluster. We propose a model of an axis-dependent ipsilateral restriction of chromosome oscillations throughout mitosis.
Collapse
|
5
|
Cai P, Casas CJ, Plancarte GQ, Hua LL, Mikawa T. Ipsilateral restriction of chromosome movement along a centrosome, and apical-basal axis during the cell cycle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.27.534352. [PMID: 37034601 PMCID: PMC10081237 DOI: 10.1101/2023.03.27.534352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Little is known about how distance between homologous chromosomes are controlled during the cell cycle. Here, we show that the distribution of centromere components display two discrete clusters placed to either side of the centrosome and apical/basal axis from prophase to G 1 interphase. 4-Dimensional live cell imaging analysis of centromere and centrosome tracking reveals that centromeres oscillate largely within one cluster, but do not cross over to the other cluster. We propose a model of an axis-dependent ipsilateral restriction of chromosome oscillations throughout mitosis.
Collapse
|
6
|
Yin Y, Zhao SL, Rane D, Lin Z, Wu M, Peterson BR. Quantification of Binding of Small Molecules to Native Proteins Overexpressed in Living Cells. J Am Chem Soc 2024; 146:187-200. [PMID: 38118119 PMCID: PMC10910633 DOI: 10.1021/jacs.3c07488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
The affinity and selectivity of small molecules for proteins drive drug discovery and development. We report a fluorescent probe cellular binding assay (FPCBA) for determination of these values for native (untagged) proteins overexpressed in living cells. This method uses fluorophores such as Pacific Blue (PB) linked to cell-permeable protein ligands to generate probes that rapidly and reversibly equilibrate with intracellular targets, as established by kinetic assays of cellular uptake and efflux. To analyze binding to untagged proteins, an internal ribosomal entry site (IRES) vector was employed that allows a single mRNA to encode both the protein target and a separate orthogonal fluorescent protein (mVenus). This enabled cellular uptake of the probe to be correlated with protein expression by flow cytometry, allowing measurement of cellular dissociation constants (Kd) of the probe. This approach was validated by studies of the binding of allosteric activators to eight different Protein Kinase C (PKC) isozymes. Full-length PKCs expressed in transiently transfected HEK293T cells were used to measure cellular Kd values of a probe comprising PB linked to the natural product phorbol via a carbamate. These values were further used to determine competitive binding constants (cellular Ki values) of the nonfluorescent phorbol ester PDBu and the anticancer agent bryostatin 1 for each isozyme. For some PKC-small molecule pairs, these cellular Ki values matched known biochemical Ki values, but for others, altered selectivity was observed in cells. This approach can facilitate quantification of interactions of small molecules with physiologically relevant native proteins.
Collapse
Affiliation(s)
- Yuwen Yin
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, College of Pharmacy, 500 W. 12 Ave., Columbus, OH 43210, USA
| | - Serena Li Zhao
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, College of Pharmacy, 500 W. 12 Ave., Columbus, OH 43210, USA
| | - Digamber Rane
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, College of Pharmacy, 500 W. 12 Ave., Columbus, OH 43210, USA
| | - Zhihong Lin
- The Ohio State University Comprehensive Cancer Center – Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, 460 W. 10 Ave., Columbus, OH 43210, USA
| | - Meng Wu
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, College of Pharmacy, 500 W. 12 Ave., Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center – Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, 460 W. 10 Ave., Columbus, OH 43210, USA
| | - Blake R. Peterson
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, College of Pharmacy, 500 W. 12 Ave., Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center – Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, 460 W. 10 Ave., Columbus, OH 43210, USA
| |
Collapse
|
7
|
Guo X, Ma Y, Wang H, Yin H, Shi X, Chen Y, Gao G, Sun L, Wang J, Wang Y, Fan D. Status and developmental trends in recombinant collagen preparation technology. Regen Biomater 2023; 11:rbad106. [PMID: 38173768 PMCID: PMC10761200 DOI: 10.1093/rb/rbad106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/20/2023] [Accepted: 11/24/2023] [Indexed: 01/05/2024] Open
Abstract
Recombinant collagen is a pivotal topic in foundational biological research and epitomizes the application of critical bioengineering technologies. These technological advancements have profound implications across diverse areas such as regenerative medicine, organ replacement, tissue engineering, cosmetics and more. Thus, recombinant collagen and its preparation methodologies rooted in genetically engineered cells mark pivotal milestones in medical product research. This article provides a comprehensive overview of the current genetic engineering technologies and methods used in the production of recombinant collagen, as well as the conventional production process and quality control detection methods for this material. Furthermore, the discussion extends to foresee the strides in physical transfection and magnetic control sorting studies, envisioning an enhanced preparation of recombinant collagen-seeded cells to further fuel recombinant collagen production.
Collapse
Affiliation(s)
- Xiaolei Guo
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing 100081, China
| | - Yuan Ma
- State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China
| | - Hang Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Hongping Yin
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Xinli Shi
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing 100081, China
| | - Yiqin Chen
- State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China
| | - Guobiao Gao
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing 100081, China
| | - Lei Sun
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing 100081, China
| | - Jiadao Wang
- State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials & College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Daidi Fan
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| |
Collapse
|
8
|
Gao Y, Wei J, Pu L, Fu S, Xing X, Zhang R, Jin F. Remotely Controllable Engineered Bacteria for Targeted Therapy of Pseudomonas aeruginosa Infection. ACS Synth Biol 2023. [PMID: 37418677 DOI: 10.1021/acssynbio.2c00655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2023]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) infection has become an intractable problem worldwide due to the decreasing efficacy of the mainstay therapy, antibiotic treatment. Hence, exploring new drugs and therapies to address this issue is crucial. Here, we construct a chimeric pyocin (ChPy) to specifically kill P. aeruginosa and engineer a near-infrared (NIR) light-responsive strain to produce and deliver this drug. Our engineered bacterial strain can continuously produce ChPy in the absence of light and release it to kill P. aeruginosa via remotely and precisely controlled bacterial lysis induced by NIR light. We demonstrate that our engineered bacterial strain is effective in P. aeruginosa-infected wound therapy in the mouse model, as it eradicated PAO1 in mouse wounds and shortened the wound healing time. Our work presents a potentially spatiotemporal and noninvasively controlled therapeutic strategy of engineered bacteria for the targeted treatment of P. aeruginosa infections.
Collapse
Affiliation(s)
- Yanmei Gao
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, No. 96, JinZhai Road, Baohe District, Hefei, Anhui 230026, P. R. China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Jingjing Wei
- Department of Fine Chemical Engineering, Shenzhen Polytechnic, Nanshan District, Shenzhen, Guangdong 518055, China
| | - Lu Pu
- West China School of Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan 610065, China
| | - Shengwei Fu
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, No. 96, JinZhai Road, Baohe District, Hefei, Anhui 230026, P. R. China
| | - Xiaochen Xing
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Rongrong Zhang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Fan Jin
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, No. 96, JinZhai Road, Baohe District, Hefei, Anhui 230026, P. R. China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| |
Collapse
|
9
|
van Zanten TS, S GP, Mayor S. Quantitative fluorescence emission anisotropy microscopy for implementing homo-fluorescence resonance energy transfer measurements in living cells. Mol Biol Cell 2023; 34:tp1. [PMID: 37144969 DOI: 10.1091/mbc.e22-09-0446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
Quantitative fluorescence emission anisotropy microscopy reveals the organization of fluorescently labeled cellular components and allows their characterization in terms of changes in either rotational diffusion or homo-Förster's energy transfer characteristics in living cells. These properties provide insights into molecular organization, such as orientation, confinement, and oligomerization in situ. Here we elucidate how quantitative measurements of anisotropy using multiple microscope systems may be made by bringing out the main parameters that influence the quantification of fluorescence emission anisotropy. We focus on a variety of parameters that contribute to errors associated with the measurement of emission anisotropy in a microscope. These include the requirement for adequate photon counts for the necessary discrimination of anisotropy values, the influence of extinction ratios of the illumination source, the detector system, the role of numerical aperture, and excitation wavelength. All these parameters also affect the ability to capture the dynamic range of emission anisotropy necessary for quantifying its reduction due to homo-FRET and other processes. Finally, we provide easily implementable tests to assess whether homo-FRET is a cause for the observed emission depolarization.
Collapse
Affiliation(s)
- Thomas S van Zanten
- Cell Biology Group, National Centre for Biological Sciences, UAS-GKVK Campus, Tata Institute for Fundamental Research, Bangalore 560065, India
| | - Greeshma Pradeep S
- Cell Biology Group, National Centre for Biological Sciences, UAS-GKVK Campus, Tata Institute for Fundamental Research, Bangalore 560065, India
| | - Satyajit Mayor
- Cell Biology Group, National Centre for Biological Sciences, UAS-GKVK Campus, Tata Institute for Fundamental Research, Bangalore 560065, India
| |
Collapse
|
10
|
Tanaka A, Kosuda M, Yamana M, Furukawa A, Nagasawa A, Fujishiro M, Kohno G, Ishihara H. A large-scale functional analysis of genes expressed differentially in insulin secreting MIN6 sublines with high versus mildly reduced glucose-responsiveness. Sci Rep 2023; 13:5654. [PMID: 37024560 PMCID: PMC10079668 DOI: 10.1038/s41598-023-32589-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 03/29/2023] [Indexed: 04/08/2023] Open
Abstract
Molecular mechanisms of glucose-stimulated insulin secretion (GSIS) from pancreatic β-cells are not fully understood. GSIS deteriorations are believed to underlie the pathogenesis of type 2 diabetes mellitus. By comparing transcript levels of 3 insulin secreting MIN6 cell sublines with strong glucose-responsiveness and 3 with mildly reduced responsiveness, we identified 630 differentially expressed genes. Using our recently developed system based on recombinase-mediated cassette exchange, we conducted large-scale generation of stable clones overexpressing such genes in the doxycycline-regulated manner. We found that overexpressions of 18, out of 83, genes altered GSIS. Sox11 ((sex determining region Y)-box 11) was selected to confirm its roles in regulating insulin secretion, and the gene was subjected to shRNA-mediated suppression. While Sox11 overexpression decreased GSIS, its suppression increased GSIS, confirming the role of Sox11 as a negative regulator of insulin secretion. Furthermore, metabolic experiments using radiolabelled glucose showed Sox11 to participate in regulating glucose metabolism. Our data suggested that overexpression screening is a feasible option for systemic functional testing to identify important genes in GSIS.
Collapse
Affiliation(s)
- Aya Tanaka
- Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, 30-1 Oyaguchikami-cho, Itabashi, 173-8610, Japan
| | - Minami Kosuda
- Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, 30-1 Oyaguchikami-cho, Itabashi, 173-8610, Japan
| | - Midori Yamana
- Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, 30-1 Oyaguchikami-cho, Itabashi, 173-8610, Japan
| | - Asami Furukawa
- Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, 30-1 Oyaguchikami-cho, Itabashi, 173-8610, Japan
| | - Akiko Nagasawa
- Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, 30-1 Oyaguchikami-cho, Itabashi, 173-8610, Japan
| | - Midori Fujishiro
- Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, 30-1 Oyaguchikami-cho, Itabashi, 173-8610, Japan
| | - Genta Kohno
- Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, 30-1 Oyaguchikami-cho, Itabashi, 173-8610, Japan
| | - Hisamitsu Ishihara
- Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, 30-1 Oyaguchikami-cho, Itabashi, 173-8610, Japan.
| |
Collapse
|
11
|
Sung BH, Weaver AM. Visualization of Exosome Release and Uptake During Cell Migration Using the Live Imaging Reporter pHluorin_M153R-CD63. Methods Mol Biol 2023; 2608:83-96. [PMID: 36653703 DOI: 10.1007/978-1-0716-2887-4_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Exosome secretion and uptake regulate cell migration through autocrine and paracrine mechanisms. Monitoring exosome secretion and uptake during cell migration is critical for investigation of these mechanisms. Exosomes can be visualized by direct labeling with fluorescent dyes or by tagging intrinsic markers with fluorescent proteins for live imaging. Due to several limitations of fluorescent dye-labeled exosomes, we created two bright genetically encoded reporters of exosome secretion, pHluorin_M153R-CD63 and pHluorin_M153R-CD63-mScarlet. Here, we describe how to visualize secretion and uptake of exosomes labeled with these pH-sensitive and pH-insensitive fluorescent protein-tagged exosomal markers during cell migration using time-lapse fluorescent microscopy.
Collapse
Affiliation(s)
- Bong H Sung
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | | |
Collapse
|
12
|
Savar NS, Vallet T, Arashkia A, Lundstrom K, Vignuzzi M, Mahmoudzadeh Niknam H. Packaging, Purification, and Titration of Replication-Deficient Semliki Forest Virus-Derived Particles as a Self-Amplifying mRNA Vaccine Vector. IRANIAN BIOMEDICAL JOURNAL 2022; 26:269-78. [PMID: 35468712 PMCID: PMC9432467 DOI: 10.52547/ibj.3535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/23/2022] [Indexed: 12/02/2022]
Abstract
Background Self-amplifying mRNA is the next-generation vaccine platform with the potential advantages in efficacy and speed of development against infectious diseases and cancer. The main aim was to present optimized and rapid methods for Semliki Forest virus (SFV)-PD self-amplifying mRNA (SAM) preparation, its packaging, and titer determination. These protocols are provided for producing and harvesting the high yields of virus replicon particle (VRP)-packaged SAM for vaccine studies. Methods pSFV-PD-EGFP plasmid was linearized and subjected to in vitro transcription. Different concentrations of SFV-PD SAM were first transfected into human embryonic kidney 293 cells (HEK-293) and baby hamster kidney cell line 21 (BHK-21) cell lines, and EGFP expression at different time points was evaluated by fluorescent microscopy. Replicon particle packaging was achieved by co-transfection of SFV-PD SAM and pSFV-Helper2 RNA into BHK-21 cells. The VRPs were concentrated using ultrafiltration with 100 kDa cut-off. The titers of replicon particles were determined by reverse transcription quantitative real-time PCR (RT-qPCR). Results In vitro transcribed SAM encoding EGFP was successfully transfected and expressed in HEK-293 and BHK-21 cell lines. Higher levels of EGFP expression was observed in BHK-21 compared to HEK-293 cells showing more stable protein overexpression and VRP packaging. Using ultrafiltration, the high yields of purified SFV-PD-EGFP particles were rapidly obtained with only minor loss of replicon particles. Accurate and rapid titer determination of replication-deficient particles was achieved by RT-qPCR. Conclusion Using optimized methods for SAM transfection, VRP packaging, and concentration, high yields of SFV-PD VRPs could be produced and purified. The RT-qPCR demonstrated to be an accurate and rapid method for titer determination of replication deficient VRPs.
Collapse
Affiliation(s)
| | - Thomas Vallet
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Arash Arashkia
- Virology Department, Pasteur Institute of Iran, Tehran, Iran
| | | | - Marco Vignuzzi
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | | |
Collapse
|
13
|
Saghaleyni R, Malm M, Moruzzi N, Zrimec J, Razavi R, Wistbacka N, Thorell H, Pintar A, Hober A, Edfors F, Chotteau V, Berggren PO, Grassi L, Zelezniak A, Svensson T, Hatton D, Nielsen J, Robinson JL, Rockberg J. Enhanced metabolism and negative regulation of ER stress support higher erythropoietin production in HEK293 cells. Cell Rep 2022; 39:110936. [PMID: 35705050 DOI: 10.1016/j.celrep.2022.110936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/05/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022] Open
Abstract
Recombinant protein production can cause severe stress on cellular metabolism, resulting in limited titer and product quality. To investigate cellular and metabolic characteristics associated with these limitations, we compare HEK293 clones producing either erythropoietin (EPO) (secretory) or GFP (non-secretory) protein at different rates. Transcriptomic and functional analyses indicate significantly higher metabolism and oxidative phosphorylation in EPO producers compared with parental and GFP cells. In addition, ribosomal genes exhibit specific expression patterns depending on the recombinant protein and the production rate. In a clone displaying a dramatically increased EPO secretion, we detect higher gene expression related to negative regulation of endoplasmic reticulum (ER) stress, including upregulation of ATF6B, which aids EPO production in a subset of clones by overexpression or small interfering RNA (siRNA) knockdown. Our results offer potential target pathways and genes for further development of the secretory power in mammalian cell factories.
Collapse
Affiliation(s)
- Rasool Saghaleyni
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Magdalena Malm
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Protein Science, 106 91 Stockholm, Sweden
| | - Noah Moruzzi
- The Rolf Luft Research Center for Diabetes and Endocrinology, Department of Molecular Medicine and Surgery, Karolinska Institute, 17176 Stockholm, Sweden
| | - Jan Zrimec
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Ronia Razavi
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Protein Science, 106 91 Stockholm, Sweden
| | - Num Wistbacka
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Protein Science, 106 91 Stockholm, Sweden
| | - Hannes Thorell
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Protein Science, 106 91 Stockholm, Sweden
| | - Anton Pintar
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Protein Science, 106 91 Stockholm, Sweden
| | - Andreas Hober
- Science for Life Laboratory, KTH - Royal Institute of Technology, 171 65 Solna, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, KTH - Royal Institute of Technology, 171 65 Solna, Sweden
| | - Veronique Chotteau
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Industrial Biotechnology, 106 91 Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Department of Molecular Medicine and Surgery, Karolinska Institute, 17176 Stockholm, Sweden
| | - Luigi Grassi
- Cell Culture & Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Aleksej Zelezniak
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Thomas Svensson
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden; Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Kemivägen 10, 41258 Gothenburg, Sweden
| | - Diane Hatton
- Cell Culture & Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Jonathan L Robinson
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden; Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Kemivägen 10, 41258 Gothenburg, Sweden.
| | - Johan Rockberg
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Department of Protein Science, 106 91 Stockholm, Sweden.
| |
Collapse
|
14
|
Zhou X, Mehta S, Zhang J. AktAR and Akt-STOPS: Genetically Encodable Molecular Tools to Visualize and Perturb Akt Kinase Activity at Different Subcellular Locations in Living Cells. Curr Protoc 2022; 2:e416. [PMID: 35532280 PMCID: PMC9093046 DOI: 10.1002/cpz1.416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The serine/threonine protein kinase Akt integrates diverse upstream inputs to regulate cell survival, growth, metabolism, migration, and differentiation. Mounting evidence suggests that Akt activity is differentially regulated depending on its subcellular location, which can include the plasma membrane, endomembrane, and nuclear compartment. This spatial control of Akt activity is critical for achieving signaling specificity and proper physiological functions, and deregulation of compartment-specific Akt signaling is implicated in various diseases, including cancer and diabetes. Understanding the spatial coordination of the signaling network centered around this key kinase and the underlying regulatory mechanisms requires precise tracking of Akt activity at distinct subcellular compartments within its native biological contexts. To address this challenge, new molecular tools are being developed, enabling us to directly interrogate the spatiotemporal regulation of Akt in living cells. These include, for instance, the newly developed genetically encodable fluorescent-protein-based Akt kinase activity reporter (AktAR2), which serves as a substrate surrogate of Akt kinase and translates Akt-specific phosphorylation into a quantifiable change in Förster resonance energy transfer (FRET). In addition, we developed the Akt substrate tandem occupancy peptide sponge (Akt-STOPS), which allows biochemical perturbation of subcellular Akt activity. Both molecular tools can be readily targeted to distinct subcellular localizations. Here, we describe a workflow to study Akt kinase activity at different subcellular locations in living cells. We provide a protocol for using genetically targeted AktAR2 and Akt-STOPS, along with fluorescence imaging in living NIH3T3 cells, to visualize and perturb, respectively, the activity of endogenous Akt kinase at different subcellular compartments. We further describe a protocol for using chemically inducible dimerization (CID) to control the plasma membrane-specific inhibition of Akt activity in real time. Lastly, we describe a protocol for maintaining NIH3T3 cells in culture, a cell line known to exhibit robust Akt activity. In all, this approach enables interrogation of spatiotemporal regulation and functions of Akt, as well as the intricate signaling networks in which it is embedded, at specific subcellular locations. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Visualizing and perturbing subcellular Akt kinase activity using AktAR and Akt-STOPS Basic Protocol 2: Using chemically inducible dimerization (CID) to control inhibition of Akt at the plasma membrane Support Protocol: Maintaining NIH3T3 cells in culture.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Pharmacology, University of California, San Diego, La Jolla, California
| | - Sohum Mehta
- Department of Pharmacology, University of California, San Diego, La Jolla, California
| | - Jin Zhang
- Department of Pharmacology, University of California, San Diego, La Jolla, California.,Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, California.,Department of Bioengineering, University of California, San Diego, La Jolla, California
| |
Collapse
|
15
|
Baedyananda F, Chaiwongkot A, Varadarajan S, Bhattarakosol P. HPV16 E1 dysregulated cellular genes involved in cell proliferation and host DNA damage: A possible role in cervical carcinogenesis. PLoS One 2021; 16:e0260841. [PMID: 34968392 PMCID: PMC8717967 DOI: 10.1371/journal.pone.0260841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 11/17/2021] [Indexed: 12/28/2022] Open
Abstract
HPV16 is the most prominent cause of cervical cancer. HPV16 E1, a helicase required for HPV replication exhibits increased expression in association with cervical cancer progression, suggesting that E1 has a similar effect on the host as the HPV16 E6 and E7 oncoproteins. This study aimed to determine whether expression of HPV16 E1 correlated with carcinogenesis by modulating cellular pathways involved in cervical cancer. HEK293T cells were transfected with pEGFP, pEGFPE1 or truncated forms of HPV16 E1. Cell proliferation, cell death, and the impact of HPV16 E1 on host gene expression was then evaluated. HPV16 E1 overexpression resulted in a significant reduction of cell viability and cellular proliferation (p-value<0.0001). Moreover, prolonged expression of HPV16 E1 significantly induced both apoptotic and necrotic cell death, which was partially inhibited by QVD-OPH, a broad-spectrum caspase inhibitor. Microarray, real time RT-PCR and kinetic host gene expression analyses revealed that HPV16 E1 overexpression resulted in the downregulation of genes involved in protein synthesis (RPL36A), metabolism (ALDOC), cellular proliferation (CREB5, HIF1A, JMJDIC, FOXO3, NFKB1, PIK3CA, TSC22D3), DNA damage (ATR, BRCA1 and CHEK1) and immune response (ISG20) pathways. How these genetic changes contribute to HPV16 E1-mediated cervical carcinogenesis warrants further studies.
Collapse
Affiliation(s)
- Fern Baedyananda
- Applied Medical Virology Research Unit, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Division of Virology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Arkom Chaiwongkot
- Applied Medical Virology Research Unit, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Division of Virology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Shankar Varadarajan
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Parvapan Bhattarakosol
- Applied Medical Virology Research Unit, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Division of Virology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
16
|
Yousefi R, Jevdokimenko K, Kluever V, Pacheu-Grau D, Fornasiero EF. Influence of Subcellular Localization and Functional State on Protein Turnover. Cells 2021; 10:cells10071747. [PMID: 34359917 PMCID: PMC8306977 DOI: 10.3390/cells10071747] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/02/2021] [Accepted: 07/07/2021] [Indexed: 12/14/2022] Open
Abstract
Protein homeostasis is an equilibrium of paramount importance that maintains cellular performance by preserving an efficient proteome. This equilibrium avoids the accumulation of potentially toxic proteins, which could lead to cellular stress and death. While the regulators of proteostasis are the machineries controlling protein production, folding and degradation, several other factors can influence this process. Here, we have considered two factors influencing protein turnover: the subcellular localization of a protein and its functional state. For this purpose, we used an imaging approach based on the pulse-labeling of 17 representative SNAP-tag constructs for measuring protein lifetimes. With this approach, we obtained precise measurements of protein turnover rates in several subcellular compartments. We also tested a selection of mutants modulating the function of three extensively studied proteins, the Ca2+ sensor calmodulin, the small GTPase Rab5a and the brain creatine kinase (CKB). Finally, we followed up on the increased lifetime observed for the constitutively active Rab5a (Q79L), and we found that its stabilization correlates with enlarged endosomes and increased interaction with membranes. Overall, our data reveal that both changes in protein localization and functional state are key modulators of protein turnover, and protein lifetime fluctuations can be considered to infer changes in cellular behavior.
Collapse
Affiliation(s)
- Roya Yousefi
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany; (R.Y.); (K.J.); (V.K.)
- Department of Cellular Biochemistry, University Medical Center Göttingen, 37073 Göttingen, Germany;
| | - Kristina Jevdokimenko
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany; (R.Y.); (K.J.); (V.K.)
| | - Verena Kluever
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany; (R.Y.); (K.J.); (V.K.)
| | - David Pacheu-Grau
- Department of Cellular Biochemistry, University Medical Center Göttingen, 37073 Göttingen, Germany;
| | - Eugenio F. Fornasiero
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany; (R.Y.); (K.J.); (V.K.)
- Correspondence:
| |
Collapse
|
17
|
Heinrich MA, Mostafa AMRH, Morton JP, Hawinkels LJAC, Prakash J. Translating complexity and heterogeneity of pancreatic tumor: 3D in vitro to in vivo models. Adv Drug Deliv Rev 2021; 174:265-293. [PMID: 33895214 DOI: 10.1016/j.addr.2021.04.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive type of cancer with an overall survival rate of less than 7-8%, emphasizing the need for novel effective therapeutics against PDAC. However only a fraction of therapeutics which seemed promising in the laboratory environment will eventually reach the clinic. One of the main reasons behind this low success rate is the complex tumor microenvironment (TME) of PDAC, a highly fibrotic and dense stroma surrounding tumor cells, which supports tumor progression as well as increases the resistance against the treatment. In particular, the growing understanding of the PDAC TME points out a different challenge in the development of efficient therapeutics - a lack of biologically relevant in vitro and in vivo models that resemble the complexity and heterogeneity of PDAC observed in patients. The purpose and scope of this review is to provide an overview of the recent developments in different in vitro and in vivo models, which aim to recapitulate the complexity of PDAC in a laboratory environment, as well to describe how 3D in vitro models can be integrated into drug development pipelines that are already including sophisticated in vivo models. Hereby a special focus will be given on the complexity of in vivo models and the challenges in vitro models face to reach the same levels of complexity in a controllable manner. First, a brief introduction of novel developments in two dimensional (2D) models and ex vivo models is provided. Next, recent developments in three dimensional (3D) in vitro models are described ranging from spheroids, organoids, scaffold models, bioprinted models to organ-on-chip models including a discussion on advantages and limitations for each model. Furthermore, we will provide a detailed overview on the current PDAC in vivo models including chemically-induced models, syngeneic and xenogeneic models, highlighting hetero- and orthotopic, patient-derived tissues (PDX) models, and genetically engineered mouse models. Finally, we will provide a discussion on overall limitations of both, in vitro and in vivo models, and discuss necessary steps to overcome these limitations to reach an efficient drug development pipeline, as well as discuss possibilities to include novel in silico models in the process.
Collapse
Affiliation(s)
- Marcel A Heinrich
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Ahmed M R H Mostafa
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Jennifer P Morton
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Rd, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Rd, Glasgow G61 1QH, UK
| | - Lukas J A C Hawinkels
- Department of Gastroenterology-Hepatology, Leiden University Medical Centre, PO-box 9600, 2300 RC Leiden, the Netherlands
| | - Jai Prakash
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands.
| |
Collapse
|
18
|
Chong ZX, Yeap SK, Ho WY. Transfection types, methods and strategies: a technical review. PeerJ 2021; 9:e11165. [PMID: 33976969 PMCID: PMC8067914 DOI: 10.7717/peerj.11165] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 03/05/2021] [Indexed: 12/17/2022] Open
Abstract
Transfection is a modern and powerful method used to insert foreign nucleic acids into eukaryotic cells. The ability to modify host cells' genetic content enables the broad application of this process in studying normal cellular processes, disease molecular mechanism and gene therapeutic effect. In this review, we summarized and compared the findings from various reported literature on the characteristics, strengths, and limitations of various transfection methods, type of transfected nucleic acids, transfection controls and approaches to assess transfection efficiency. With the vast choices of approaches available, we hope that this review will help researchers, especially those new to the field, in their decision making over the transfection protocol or strategy appropriate for their experimental aims.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- School of Pharmacy, University of Nottingham Malaysia, Semenyih, Selangor, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Sepang, Selangor, Malaysia
| | - Wan Yong Ho
- School of Pharmacy, University of Nottingham Malaysia, Semenyih, Selangor, Malaysia
| |
Collapse
|
19
|
Kuo JCH, Goudge MC, Metzloff AE, Huang LT, Colville MJ, Park S, Zipfel WR, Paszek MJ. Litmus-Body: A Molecularly Targeted Sensor for Cell-Surface pH Measurements. ACS Sens 2020; 5:1555-1566. [PMID: 32337979 PMCID: PMC8858598 DOI: 10.1021/acssensors.9b02080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Precise pH measurements in the immediate environment of receptors is essential for elucidating the mechanisms through which local pH changes associated with diseased phenotypes manifest into aberrant receptor function. However, current pH sensors lack the ability to localize and target specific receptor molecules required to make these measurements. Herein we present the Litmus-body, our recombinant protein-based pH sensor, which through fusion to an anti-IgG nanobody is capable of piggybacking on IgG antibodies for molecular targeting to specific proteins on the cell surface. By normalizing a pH-dependent green fluorescent protein to a long Stokes shift red fluorophore or fluorescent protein, we readily report pH independent of sensor concentration using a single 488 nm excitation. Our Litmus-body showed excellent responsiveness in solution, with a greater than 50-fold change across the regime of physiological pH. The sensor was further validated for use on live cells and shown to be specific to the protein of interest. In complex with our Litmus-body, cetuximab therapeutic antibody retained its functionality in binding and inhibiting ligand interaction of its target epidermal growth factor receptor (EGFR), triggering receptor-mediated endocytosis that allowed tracking of local pH from the cell surface through the endocytic pathway.
Collapse
Affiliation(s)
- Joe Chin-Hun Kuo
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Marc C. Goudge
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Ann E. Metzloff
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Ling-Ting Huang
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | - Sangwoo Park
- Field of Biophysics, Cornell University, Ithaca, NY 14853, USA
| | - Warren R. Zipfel
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Field of Biophysics, Cornell University, Ithaca, NY 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Ithaca, NY 14853, USA
| | - Matthew J. Paszek
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Field of Biophysics, Cornell University, Ithaca, NY 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Ithaca, NY 14853, USA
| |
Collapse
|