1
|
Pattanakittivorakul S, Kato S, Kuga T, Kosaka T, Matsutani M, Murata M, Ishikawa M, Charoenpunthuwong K, Thanonkeo P, Yamada M. Limited Diversity of Thermal Adaptation to a Critical Temperature in Zymomonas mobilis: Evidence from Multiple-Parallel Laboratory Evolution Experiments. Int J Mol Sci 2025; 26:3052. [PMID: 40243698 PMCID: PMC11989028 DOI: 10.3390/ijms26073052] [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: 02/08/2025] [Revised: 03/07/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Laboratory evolution is an effective means of understanding microbial adaptation to the environment. We previously isolated four thermoadapted Zymomonas mobilis mutants, which showed a 2 °C rise in the critical high temperature (CHT), by performing multiple-parallel adaptation experiments. In the present study, the individual mutations in these mutants were intensively analyzed. Two mutations in each adapted mutant were found to primarily contribute to the increase in the upper temperature limit. RNA sequencing (RNA-seq) analysis revealed that the two mutations led to the upregulation of 79-185 genes and the downregulation of 242-311 genes. The findings from transcriptomic and physiological experiments suggest two common and primary mechanisms for thermal resistance: a decrease in the activity of diacylglycerol kinase, which may change the structure of lipopolysaccharide (LPS) probably to strengthen the membrane structure, and an increase in the expression of genes for GroEL/GroES or cell wall hydrolase to repair the protein or membrane damage that occurs at such critical temperatures. Additionally, transporters including efflux pumps may contribute to intracellular homeostasis by expelling toxic compounds such as ethanol and acetate or by maintaining the K+ concentration. The results of this study on four independently thermoadapted mutants led to the conclusion that the mutants have almost the same thermal adaptation strategies and thus their molecular diversity is limited.
Collapse
Affiliation(s)
- Sornsiri Pattanakittivorakul
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8515, Japan; (S.P.); (S.K.); (T.K.); (T.K.); (M.M.)
| | - Shun Kato
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8515, Japan; (S.P.); (S.K.); (T.K.); (T.K.); (M.M.)
| | - Takashi Kuga
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8515, Japan; (S.P.); (S.K.); (T.K.); (T.K.); (M.M.)
| | - Tomoyuki Kosaka
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8515, Japan; (S.P.); (S.K.); (T.K.); (T.K.); (M.M.)
- Research Center for Thermotolerant Microbial Resources, Yamaguchi University, Yamaguchi 753-8515, Japan
| | - Minenosuke Matsutani
- NODAI Genome Research Center, Tokyo University of Agriculture, Tokyo 156-8502, Japan;
| | - Masayuki Murata
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8515, Japan; (S.P.); (S.K.); (T.K.); (T.K.); (M.M.)
| | - Morio Ishikawa
- Department of Bioscience, Tokyo University of Agriculture, Tokyo 156-8502, Japan;
| | - Kankanok Charoenpunthuwong
- Department of Biotechnology, Faculty of Technology, Khon Kaen University, Khon Kaen 40002, Thailand; (K.C.); (P.T.)
| | - Pornthap Thanonkeo
- Department of Biotechnology, Faculty of Technology, Khon Kaen University, Khon Kaen 40002, Thailand; (K.C.); (P.T.)
- Fermentation Research Center for Value Added Agricultural Products, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Mamoru Yamada
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8515, Japan; (S.P.); (S.K.); (T.K.); (T.K.); (M.M.)
- Research Center for Thermotolerant Microbial Resources, Yamaguchi University, Yamaguchi 753-8515, Japan
| |
Collapse
|
2
|
Yau CN, Hung JTS, Campbell RAA, Wong TCY, Huang B, Wong BTY, Chow NKN, Zhang L, Tsoi EPL, Tan Y, Li JJX, Wing YK, Lai HM. INSIHGT: an accessible multi-scale, multi-modal 3D spatial biology platform. Nat Commun 2024; 15:10888. [PMID: 39738072 PMCID: PMC11685604 DOI: 10.1038/s41467-024-55248-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 12/06/2024] [Indexed: 01/01/2025] Open
Abstract
Biological systems are complex, encompassing intertwined spatial, molecular and functional features. However, methodological constraints limit the completeness of information that can be extracted. Here, we report the development of INSIHGT, a non-destructive, accessible three-dimensional (3D) spatial biology method utilizing superchaotropes and host-guest chemistry to achieve homogeneous, deep penetration of macromolecular probes up to centimeter scales, providing reliable semi-quantitative signals throughout the tissue volume. Diverse antigens, mRNAs, neurotransmitters, and post-translational modifications are well-preserved and simultaneously visualized. INSIHGT also allows multi-round, highly multiplexed 3D molecular probing and is compatible with downstream traditional histology and nucleic acid sequencing. With INSIHGT, we map undescribed podocyte-to-parietal epithelial cell microfilaments in mouse glomeruli and neurofilament-intensive inclusion bodies in the human cerebellum, and identify NPY-proximal cell types defined by spatial morpho-proteomics in mouse hypothalamus. We anticipate that INSIHGT can form the foundations for 3D spatial multi-omics technology development and holistic systems biology studies.
Collapse
Affiliation(s)
- Chun Ngo Yau
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jacky Tin Shing Hung
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Robert A A Campbell
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK
| | - Thomas Chun Yip Wong
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bei Huang
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ben Tin Yan Wong
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Nick King Ngai Chow
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lichun Zhang
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Eldric Pui Lam Tsoi
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuqi Tan
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Joshua Jing Xi Li
- Department of Pathology, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - Yun Kwok Wing
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hei Ming Lai
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Department of Chemical Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
3
|
Ma FH, Li C, Liu Y, Shi L. Mimicking Molecular Chaperones to Regulate Protein Folding. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1805945. [PMID: 31045287 DOI: 10.1002/adma.201805945] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/28/2019] [Indexed: 06/09/2023]
Abstract
Folding and unfolding are essential ways for a protein to regulate its biological activity. The misfolding of proteins usually reduces or completely compromises their biological functions, which eventually causes a wide range of diseases including neurodegeneration diseases, type II diabetes, and cancers. Therefore, materials that can regulate protein folding and maintain proteostasis are of significant biological and medical importance. In living organisms, molecular chaperones are a family of proteins that maintain proteostasis by interacting with, stabilizing, and repairing various non-native proteins. In the past few decades, efforts have been made to create artificial systems to mimic the structure and biological functions of nature chaperonins. Herein, recent progress in the design and construction of materials that mimic different kinds of natural molecular chaperones is summarized. The fabrication methods, construction rules, and working mechanisms of these artificial chaperone systems are described. The application of these materials in enhancing the thermal stability of proteins, assisting de novo folding of proteins, and preventing formation of toxic protein aggregates is also highlighted and explored. Finally, the challenges and potential in the field of chaperone-mimetic materials are discussed.
Collapse
Affiliation(s)
- Fei-He Ma
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Chang Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Yang Liu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
4
|
Wang H, Yang Y, Xu J, Kong D, Li Y. iTRAQ-based comparative proteomic analysis of differentially expressed proteins in Rhodococcus sp. BAP-1 induced by fluoranthene. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 169:282-291. [PMID: 30458394 DOI: 10.1016/j.ecoenv.2018.11.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/16/2018] [Accepted: 11/06/2018] [Indexed: 06/09/2023]
Abstract
To reveal the molecular mechanism at the level of regulation of proteins in Rhodococcus sp. BAP-1 induced by fluoranthene comparative proteomic analysis was performed on proteins extracted from fluoranthene-exposed cells on 1 d, 3 d, 6 d and 8 d compared with control cells using isobaric tags for relative and absolute quantization (iTRAQ) labeling and LC-MS/MS analysis to access differentially expressed proteins. As a result, we detected a total of 897 significantly differentially expressed proteins, including 30 shared proteins in four comparison clusters. We were able to short-list 190, 329, 101 and 90 proteins that were over-represented, and 394, 234, 65 and 49 under-represented proteins, in 1d/control, 3d/control, 6d/control and 8d/control comparisons, respectively. Functional analysis relied on Clusters of Orthologous Groups (COG), gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) revealed that fluoranthene significantly altered the expression of proteins involved in metabolic and biosynthesis processes. Furthermore, BAP-1 up-regulates aldehyde dehydrogenase, cytochrome c oxidase, and oligopeptide transport ATP-binding protein, while down-regulates several other proteins in order to adapt to fluoranthene exposure. These findings provide important clues to reveal fluoranthene degradation mechanism in BAP-1 and promote its bioremediation applications.
Collapse
Affiliation(s)
- Hongqi Wang
- College of Water Sciences, Beijing Normal University, 100875 Beijing, China
| | - Yan Yang
- College of Water Sciences, Beijing Normal University, 100875 Beijing, China
| | - Jie Xu
- College of Water Sciences, Beijing Normal University, 100875 Beijing, China
| | - Dekang Kong
- College of Water Sciences, Beijing Normal University, 100875 Beijing, China
| | - Yi Li
- College of Environment and Resource, Guangxi Normal University, 541004 Guilin, Guangxi, China.
| |
Collapse
|
5
|
Dahiya V, Buchner J. Functional principles and regulation of molecular chaperones. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 114:1-60. [PMID: 30635079 DOI: 10.1016/bs.apcsb.2018.10.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
To be able to perform their biological function, a protein needs to be correctly folded into its three dimensional structure. The protein folding process is spontaneous and does not require the input of energy. However, in the crowded cellular environment where there is high risk of inter-molecular interactions that may lead to protein molecules sticking to each other, hence forming aggregates, protein folding is assisted. Cells have evolved robust machinery called molecular chaperones to deal with the protein folding problem and to maintain proteins in their functional state. Molecular chaperones promote efficient folding of newly synthesized proteins, prevent their aggregation and ensure protein homeostasis in cells. There are different classes of molecular chaperones functioning in a complex interplay. In this review, we discuss the principal characteristics of different classes of molecular chaperones, their structure-function relationships, their mode of regulation and their involvement in human disorders.
Collapse
Affiliation(s)
- Vinay Dahiya
- Center for Integrated Protein Science Munich CIPSM at the Department Chemie, Technische Universität München, Garching, Germany
| | - Johannes Buchner
- Center for Integrated Protein Science Munich CIPSM at the Department Chemie, Technische Universität München, Garching, Germany.
| |
Collapse
|
6
|
GroEL/ES mediated the in vivo recovery of TRAIL inclusion bodies in Escherichia coli. Sci Rep 2018; 8:15766. [PMID: 30361617 PMCID: PMC6202318 DOI: 10.1038/s41598-018-34090-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 09/18/2018] [Indexed: 01/01/2023] Open
Abstract
Inclusion body (IB) formation generates substantial bio-waste in the pharmaceutical industry and remains a major challenge for heterologous protein expression. Although chaperones can be co-expressed to improve soluble protein yield, their contribution to IB processing in vivo has not been thoroughly studied. Here, a GroEL-GroES co-expressing strain and a deficient strain were constructed to study the in vivo recovery of recombinant human tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). The interaction between GroEL/ES and TRAIL was simulated by molecular docking and identified by co-immunoprecipitation. The in vitro cytotoxicity of TRAIL IBs before and after in vivo recovery was subsequently determined by MTT assay. Additionally, IB structures were measured by Fourier transform infrared (FT-IR) spectroscopy and fluorescence spectroscopy. The results showed that after in vivo refolding, IBs retained lower levels of anti-tumor activity and fewer native-like β-sheet structures. Fewer recoverable polypeptides were trapped in IBs after GroEL/ES co-expression and refolding in vivo. Therefore, GroEL/ES mediated the in vivo recovery of TRAIL IBs in Escherichia coli. These results may identify potential uses for IBs and provide additional insight into the detailed mechanisms of in vivo protein recovery.
Collapse
|
7
|
Kumar V, Chaudhuri TK. Spontaneous refolding of the large multidomain protein malate synthase G proceeds through misfolding traps. J Biol Chem 2018; 293:13270-13283. [PMID: 29959230 DOI: 10.1074/jbc.ra118.003903] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/28/2018] [Indexed: 11/06/2022] Open
Abstract
Most protein folding studies until now focus on single domain or truncated proteins. Although great insights in the folding of such systems has been accumulated, very little is known regarding the proteins containing multiple domains. It has been shown that the high stability of domains, in conjunction with inter-domain interactions, manifests as a frustrated energy landscape, causing complexity in the global folding pathway. However, multidomain proteins despite containing independently foldable, loosely cooperative sections can fold into native states with amazing speed and accuracy. To understand the complexity in mechanism, studies were conducted previously on the multidomain protein malate synthase G (MSG), an enzyme of the glyoxylate pathway with four distinct and adjacent domains. It was shown that the protein refolds to a functionally active intermediate state at a fast rate, which slowly produces the native state. Although experiments decoded the nature of the intermediate, a full description of the folding pathway was not elucidated. In this study, we use a battery of biophysical techniques to examine the protein's folding pathway. By using multiprobe kinetics studies and comparison with the equilibrium behavior of protein against urea, we demonstrate that the unfolded polypeptide undergoes conformational compaction to a misfolded intermediate within milliseconds of refolding. The misfolded product appears to be stabilized under moderate denaturant concentrations. Further folding of the protein produces a stable intermediate, which undergoes partial unfolding-assisted large segmental rearrangements to achieve the native state. This study reveals an evolved folding pathway of the multidomain protein MSG, which involves surpassing the multiple misfolding traps during refolding.
Collapse
Affiliation(s)
- Vipul Kumar
- From the Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi 110016, India
| | - Tapan K Chaudhuri
- From the Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi 110016, India
| |
Collapse
|
8
|
Molecular Chaperones: Structure-Function Relationship and their Role in Protein Folding. REGULATION OF HEAT SHOCK PROTEIN RESPONSES 2018. [DOI: 10.1007/978-3-319-74715-6_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
9
|
Weiss C, Jebara F, Nisemblat S, Azem A. Dynamic Complexes in the Chaperonin-Mediated Protein Folding Cycle. Front Mol Biosci 2016; 3:80. [PMID: 28008398 PMCID: PMC5143341 DOI: 10.3389/fmolb.2016.00080] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 11/23/2016] [Indexed: 11/13/2022] Open
Abstract
The GroEL–GroES chaperonin system is probably one of the most studied chaperone systems at the level of the molecular mechanism. Since the first reports of a bacterial gene involved in phage morphogenesis in 1972, these proteins have stimulated intensive research for over 40 years. During this time, detailed structural and functional studies have yielded constantly evolving concepts of the chaperonin mechanism of action. Despite of almost three decades of research on this oligomeric protein, certain aspects of its function remain controversial. In this review, we highlight one central aspect of its function, namely, the active intermediates of its reaction cycle, and present how research to this day continues to change our understanding of chaperonin-mediated protein folding.
Collapse
Affiliation(s)
- Celeste Weiss
- George S. Weiss Faculty of Life Sciences, Department of Biochemistry and Molecular Biology, Tel Aviv University Tel Aviv, Israel
| | - Fady Jebara
- George S. Weiss Faculty of Life Sciences, Department of Biochemistry and Molecular Biology, Tel Aviv University Tel Aviv, Israel
| | - Shahar Nisemblat
- George S. Weiss Faculty of Life Sciences, Department of Biochemistry and Molecular Biology, Tel Aviv University Tel Aviv, Israel
| | - Abdussalam Azem
- George S. Weiss Faculty of Life Sciences, Department of Biochemistry and Molecular Biology, Tel Aviv University Tel Aviv, Israel
| |
Collapse
|
10
|
Effect of culture conditions on the whole cell activity of recombinant Escherichia coli expressing periplasmic organophosphorus hydrolase and cytosolic GroEL/ES chaperone. BIOTECHNOL BIOPROC E 2016. [DOI: 10.1007/s12257-016-0342-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
11
|
Interaction of Mycobacterium tuberculosis Virulence Factor RipA with Chaperone MoxR1 Is Required for Transport through the TAT Secretion System. mBio 2016; 7:e02259. [PMID: 26933057 PMCID: PMC4810496 DOI: 10.1128/mbio.02259-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mycobacterium tuberculosis is a leading cause of death worldwide. The M. tuberculosis TAT (twin-arginine translocation) protein secretion system is present at the cytoplasmic membrane of mycobacteria and is known to transport folded proteins. The TAT secretion system is reported to be essential for many important bacterial processes that include cell wall biosynthesis. The M. tuberculosis secretion and invasion protein RipA has endopeptidase activity and interacts with one of the resuscitation antigens (RpfB) that are expressed during pathogen reactivation. MoxR1, a member of the ATPase family that is associated with various cellular activities, was predicted to interact with RipA based on in silico analyses. A bimolecular fluorescence complementation (BiFC) assay confirmed the interaction of these two proteins in HEK293T cells. The overexpression of RipA in Mycobacterium smegmatis and copurification with MoxR1 further validated their interaction in vivo. Recombinant MoxR1 protein, expressed in Escherichia coli, displays ATP-enhanced chaperone activity. Secretion of recombinant RipA (rRipA) protein into the E. coli culture filtrate was not observed in the absence of RipA-MoxR interaction. Inhibition of this export system in M. tuberculosis, including the key players, will prevent localization of peptidoglycan hydrolase and result in sensitivity to existing β-lactam antibiotics, opening up new candidates for drug repurposing. The virulence mechanism of mycobacteria is very complex. Broadly, the virulence factors can be classified as secretion factors, cell surface components, enzymes involved in cellular metabolism, and transcriptional regulators. The mycobacteria have evolved several mechanisms to secrete its proteins. Here, we have identified one of the virulence proteins of Mycobacterium tuberculosis, RipA, possessing peptidoglycan hydrolase activities secreted by the TAT secretion pathway. We also identified MoxR1 as a protein-protein interaction partner of RipA and demonstrated chaperone activity of this protein. We show that MoxR1-mediated folding is critical for the secretion of RipA within the TAT system. Inhibition of this export system in M. tuberculosis will prevent localization of peptidoglycan hydrolase and result in sensitivity to existing β-lactam antibiotics, opening up new candidates for drug repurposing.
Collapse
|
12
|
Fang X, Yang T, Wang L, Yu J, Wei X, Zhou Y, Wang C, Liang W. Nano-cage-mediated refolding of insulin by PEG-PE micelle. Biomaterials 2015; 77:139-48. [PMID: 26595505 DOI: 10.1016/j.biomaterials.2015.11.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 11/05/2015] [Accepted: 11/06/2015] [Indexed: 10/22/2022]
Abstract
Insulin aggregation has pronounced pharmaceutical implications and biological importance. Deposition of insulin aggregates is associated with type II diabetes and instability of pharmaceutical formulations. We present in this study the renaturation effect of PEG-PE micelle on dithiothreitol (DTT)-denatured insulin revealed by techniques including turbidity assay, circular dichroism (CD), thioflavinT (ThT) binding assay, bis-ANS binding assay, agarose gel electrophoresis and MALDI-TOF MS. The obtained results show that PEG-PE micelle having a hydrophilic nano-cage-like structure in which with a negative charge layer, can capture DTT-induced insulin A and B chains, and block their hydrophobic interaction, thereby preventing aggregation. The reduced insulin A and B chain in the nano-cage are capable of recognizing each other and form the native insulin with yields of ∼30% as measured by hypoglycemic activity analysis in mice. The observed insulin refolding assisted by PEG-PE micelle may be applicable to other proteins.
Collapse
Affiliation(s)
- Xiaocui Fang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China; CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No.11 ZhongGuanCun BeiYiTiao, Beijing 100190, China
| | - Tao Yang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China; University of the Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Luoyang Wang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China; University of the Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Jibing Yu
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China
| | - Xiuli Wei
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China
| | - Yinjian Zhou
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China
| | - Chen Wang
- CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No.11 ZhongGuanCun BeiYiTiao, Beijing 100190, China.
| | - Wei Liang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China.
| |
Collapse
|
13
|
The GroEL-GroES Chaperonin Machine: A Nano-Cage for Protein Folding. Trends Biochem Sci 2015; 41:62-76. [PMID: 26422689 DOI: 10.1016/j.tibs.2015.07.009] [Citation(s) in RCA: 273] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 07/21/2015] [Accepted: 07/31/2015] [Indexed: 01/12/2023]
Abstract
The bacterial chaperonin GroEL and its cofactor GroES constitute the paradigmatic molecular machine of protein folding. GroEL is a large double-ring cylinder with ATPase activity that binds non-native substrate protein (SP) via hydrophobic residues exposed towards the ring center. Binding of the lid-shaped GroES to GroEL displaces the bound protein into an enlarged chamber, allowing folding to occur unimpaired by aggregation. GroES and SP undergo cycles of binding and release, regulated allosterically by the GroEL ATPase. Recent structural and functional studies are providing insights into how the physical environment of the chaperonin cage actively promotes protein folding, in addition to preventing aggregation. Here, we review different models of chaperonin action and discuss issues of current debate.
Collapse
|
14
|
Kim HW, Wi AR, Jeon BW, Lee JH, Shin SC, Park H, Jeon SJ. Cold adaptation of a psychrophilic chaperonin from Psychrobacter sp. and its application for heterologous protein expression. Biotechnol Lett 2015; 37:1887-93. [PMID: 26003095 DOI: 10.1007/s10529-015-1860-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/14/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVES A chaperonin, PsyGroELS, from the Antarctic psychrophilic bacterium Psychrobacter sp. PAMC21119, was examined for its role in cold adaptation when expressed in a mesophilic Escherichia coli strain. RESULTS Growth of E. coli harboring PsyGroELS at 10 °C was increased compared to the control strain. A co-expression system using PsyGroELS was developed to increase productivity of the psychrophilic enzyme PsyEst9. PsyEst9 was cloned and expressed using three E. coli variants that co-expressed GroELS from PAMC21119, E. coli, or Oleispira antarctica RB8(T). Co-expression with PsyGroELS was more effective for the production of PsyEst9 compared tothe other chaperonins. CONCLUSION PsyGroELS confers cold tolerance to E. coli, and shows potential as an effective co-expression system for the stable production of psychrophilic proteins.
Collapse
Affiliation(s)
- Han-Woo Kim
- Division of Polar Life Sciences, Korea Polar Research Institute (KOPRI), 26, Songdomirae-ro, Yeonsu-gu, Incheon, 406-840, Republic of Korea,
| | | | | | | | | | | | | |
Collapse
|
15
|
Gutiérrez-Estrada A, Ramírez-Santos J, Gómez-Eichelmann MDC. Role of chaperones and ATP synthase in DNA gyrase reactivation in Escherichia coli stationary-phase cells after nutrient addition. SPRINGERPLUS 2014; 3:656. [PMID: 25485196 PMCID: PMC4230433 DOI: 10.1186/2193-1801-3-656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 10/29/2014] [Indexed: 12/03/2022]
Abstract
Escherichia coli stationary-phase (SP) cells contain relaxed DNA molecules and recover DNA supercoiling once nutrients become available. In these cells, the reactivation of DNA gyrase, which is a DNA topoisomerase type IIA enzyme, is responsible for the recovery of DNA supercoiling. The results presented in this study show that DNA gyrase reactivation does not require cellular chaperones or polyphosphate. Glucose addition to SP cells induced a slow recovery of DNA supercoiling, whereas resveratrol, which is an inhibitor of ATP synthase, inhibited the enzyme reactivation. These results suggest that DNA gyrase, which is an ATP-dependent enzyme, remains soluble in SP cells, and that its reactivation occurs primarily due to a rapid increase in the cellular ATP concentration.
Collapse
Affiliation(s)
- Alejandra Gutiérrez-Estrada
- Department of Molecular Biology and Biotechnology, Institute of Biomedical Research, National Autonomous University of México, P.O. Box 70228, México City, 04510 México
| | - Jesús Ramírez-Santos
- Department of Molecular Biology and Biotechnology, Institute of Biomedical Research, National Autonomous University of México, P.O. Box 70228, México City, 04510 México
| | - María Del Carmen Gómez-Eichelmann
- Department of Molecular Biology and Biotechnology, Institute of Biomedical Research, National Autonomous University of México, P.O. Box 70228, México City, 04510 México
| |
Collapse
|
16
|
Yang Y, Huang L, Wang J, Xu Z. Expression, characterization and mutagenesis of an FAD-dependent glucose dehydrogenase from Aspergillus terreus. Enzyme Microb Technol 2014; 68:43-9. [PMID: 25435504 DOI: 10.1016/j.enzmictec.2014.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/09/2014] [Accepted: 10/14/2014] [Indexed: 11/18/2022]
Abstract
An FAD-dependent glucose dehydrogenase (FAD-GDH) from Aspergillus terreus NIH2624 was expressed in Escherichia coli with a yield of 228±16U/L of culture. Co-expression with chaperones DnaK/DnaJ/GrpE and osmotic stress induced by simple carbon sources enhanced productivity significantly, improving the yield to 23883±563U/L after optimization. FAD-GDH was purified in two steps with the specific activity of 604U/mg. Using d-glucose as substrate, the optimal pH and temperature for FAD-GDH were determined to be 7.5 and 50°C, respectively. Activity was stable across the pH range 3.5-9.0, and the half-life was 52min at 42°C. Km and Vmax were calculated as 86.7±5.3mM and 928±35U/mg, and the molecular weight was approximately 65.6kDa based on size exclusion chromatography, indicating a monomeric structure. The 3D structure of FAD-GDH was simulated by homology modelling using the structure of A. niger glucose oxidase (GOD) as template. From the model, His551, His508, Asn506 and Arg504 were identified as key residues, and their importance was verified by site-directed mutagenesis. Furthermore, three additional mutants (Arg84Ala, Tyr340Phe and Tyr406Phe) were generated and all exhibited a higher degree of substrate specificity than the native enzyme. These results extend our understanding of the structure and function of FAD-GDH, and could assist potential commercial applications.
Collapse
Affiliation(s)
- Yufeng Yang
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, PR China; Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China; Zunyi Medical College (Zhuhai Campus), Zhuhai, 519041, PR China
| | - Lei Huang
- Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Jufang Wang
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, PR China.
| | - Zhinan Xu
- Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China.
| |
Collapse
|
17
|
Lim NCH, Jackson SE. Mechanistic insights into the folding of knotted proteins in vitro and in vivo. J Mol Biol 2014; 427:248-58. [PMID: 25234087 DOI: 10.1016/j.jmb.2014.09.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/05/2014] [Accepted: 09/06/2014] [Indexed: 10/24/2022]
Abstract
The importance of knots and entanglements in biological systems is increasingly being realized and the number of proteins with topologically complex knotted structures has risen. However, the mechanism as to how these proteins knot and fold efficiently remains unclear. Using a cell-free expression system and pulse-proteolysis experiments, we have investigated the mechanism of knotting and folding for two bacterial trefoil-knotted methyltransferases. This study provides the first experimental evidence for a knotting mechanism. Results on fusions of stable protein domains to N-terminus, C-terminus or both termini of the knotted proteins clearly demonstrate that threading of the nascent chain through a knotting loop occurs via the C-terminus. Our results strongly suggest that this mechanism occurs even when the C-terminus is severely hindered by the addition of a large stable structure, in contrast to some simulations indicating that even the folding pathways of knotted proteins have some plasticity. The same strategy was employed to probe the effects of GroEL-GroES. In this case, results suggest active mechanisms for the molecular chaperonin. We demonstrate that a simple model in which GroEL-GroES sterically confines the unknotted polypeptide chain thereby promoting knotting is unlikely, and we propose two alternatives: (a) the chaperonin facilitates unfolding of kinetically and topologically trapped intermediates or (b) the chaperonin stabilizes interactions that promote knotting. These findings provide mechanistic insights into the folding of knotted proteins both in vitro and in vivo, thus elucidating how they have withstood evolutionary pressures regardless of their complex topologies and intrinsically slow folding rates.
Collapse
Affiliation(s)
- Nicole C H Lim
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom; Faculty of Sciences, Universiti Brunei Darussalam, Gadong BE 1410, Brunei Darussalam
| | - Sophie E Jackson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom.
| |
Collapse
|