1
|
Park H, Youn B, Park DJ, Puthanveettil SV, Kang C. Functional implication of the homotrimeric multidomain vacuolar sorting receptor 1 (VSR1) from Arabidopsis thaliana. Sci Rep 2024; 14:9622. [PMID: 38671060 PMCID: PMC11052993 DOI: 10.1038/s41598-024-57975-2] [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: 01/13/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
The vacuolar sorting receptors (VSRs) are specific to plants and are responsible for sorting and transporting particular proteins from the trans-Golgi network to the vacuole. This process is critically important for various cellular functions, including storing nutrients during seed development. Despite many years of intense studies on VSRs, a complete relation between function and structure has not yet been revealed. Here, we present the crystal structure of the entire luminal region of glycosylated VSR1 from Arabidopsis thaliana (AtVSR1) for the first time. The structure provides insights into the tertiary and quaternary structures of VSR1, which are composed of an N-terminal protease-associated (PA) domain, a unique central region, and one epidermal growth factor (EGF)-like domain followed by two disordered EGF-like domains. The structure of VSR1 exhibits unique characteristics, the significance of which is yet to be fully understood.
Collapse
Affiliation(s)
- HaJeung Park
- X-Ray Core, UF Scripps Biomedical Research, University of Florida, Jupiter, FL, 33458, USA
| | - BuHyun Youn
- Department of Biological Sciences, Pusan National University, Busan, 46241, Republic of Korea
| | - Daniel J Park
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA
| | | | - ChulHee Kang
- Department of Chemistry, Washington State University, Pullman, WA, 99164, USA.
| |
Collapse
|
2
|
Tan J, Xu Y, Wang X, Yan F, Xian W, Liu X, Chen Y, Zhu Y, Zhou Y. Molecular basis of threonine ADP-ribosylation of ubiquitin by bacterial ARTs. Nat Chem Biol 2024; 20:463-472. [PMID: 37945894 DOI: 10.1038/s41589-023-01475-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 10/08/2023] [Indexed: 11/12/2023]
Abstract
Ubiquitination plays essential roles in eukaryotic cellular processes. The effector protein CteC from Chromobacterium violaceum blocks host ubiquitination by mono-ADP-ribosylation of ubiquitin (Ub) at residue T66. However, the structural basis for this modification is unknown. Here we report three crystal structures of CteC in complexes with Ub, NAD+ or ADP-ribosylated Ub, which represent different catalytic states of CteC in the modification. CteC adopts a special 'D-E' catalytic motif for catalysis and binds NAD+ in a half-ligand binding mode. The specific recognition of Ub by CteC is determined by a relatively separate Ub-targeting domain and a long loop L6, not the classic ADP-ribosylating turn-turn loop. Structural analyses with biochemical results reveal that CteC represents a large family of poly (ADP-ribose) polymerase (PARP)-like ADP-ribosyltransferases, which harbors chimeric features from the R-S-E and H-Y-E classes of ADP-ribosyltransferases. The family of CteC-like ADP-ribosyltransferases has a common 'D-E' catalytic consensus and exists extensively in bacteria and eukaryotic microorganisms.
Collapse
Affiliation(s)
- Jiaxing Tan
- The MOE Key Laboratory for Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Department of Gastroenterology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yan Xu
- The MOE Key Laboratory for Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Xiaofei Wang
- The MOE Key Laboratory for Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Department of Gastroenterology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fujie Yan
- The MOE Key Laboratory for Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Wei Xian
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaoyun Liu
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yan Chen
- Department of Gastroenterology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongqun Zhu
- The MOE Key Laboratory for Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Department of Gastroenterology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yan Zhou
- Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, China.
- Department of Infectious Diseases, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
3
|
Brangulis K, Akopjana I, Bogans J, Kazaks A, Tars K. Structural studies of chromosomally encoded outer surface lipoprotein BB0158 from Borrelia burgdorferi sensu stricto. Ticks Tick Borne Dis 2024; 15:102287. [PMID: 38016210 DOI: 10.1016/j.ttbdis.2023.102287] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023]
Abstract
Lyme disease, or also known as Lyme borreliosis, is caused by the spirochetes belonging to the Borrelia burgdorferi sensu lato complex, which can enter the human body following the bite of an infected tick. Many membrane lipid-bound proteins, also known as lipoproteins, are located on the surface of B. burgdorferi sensu lato and play a crucial role in the spirochete to interact with its environment, whether in ticks or mammals. Since the spirochete needs to perform various tasks, such as resisting the host's immune system or spreading throughout the organism, it is not surprising that numerous surface proteins have been found to be essential for B. burgdorferi sensu lato complex bacteria in causing Lyme disease. In this study, we have determined (at 2.4 Å resolution) and characterized the 3D structure of BB0158, one of the few chromosomally encoded outer surface proteins from B. burgdorferi sensu stricto. BB0158 belongs to the paralogous gene family 44 (PFam44), consisting of four other members (BB0159, BBA04, BBE09 and BBK52). The characterization of BB0158, which appears to form a domain-swapped dimer, in conjunction with the characterization of the corresponding PFam44 members, certainly contribute to our understanding of B. burgdorferi sensu stricto proteins.
Collapse
Affiliation(s)
- Kalvis Brangulis
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga LV-1067, Latvia.
| | - Inara Akopjana
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga LV-1067, Latvia
| | - Janis Bogans
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga LV-1067, Latvia
| | - Andris Kazaks
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga LV-1067, Latvia
| | - Kaspars Tars
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga LV-1067, Latvia
| |
Collapse
|
4
|
Zhang T, Dong S, Zhai Y, Naatz L, Zhou Z, Chen M. Diphtheria toxin-derived, anti-PD-1 immunotoxin, a potent and practical tool to selectively deplete PD-1 + cells. Protein Sci 2023; 32:e4741. [PMID: 37515422 PMCID: PMC10443333 DOI: 10.1002/pro.4741] [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: 02/04/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 07/30/2023]
Abstract
Programmed death-1 (PD-1), an immune checkpoint receptor, is expressed on activated lymphocytes, macrophages, and some types of tumor cells. While PD-1+ cells have been implicated in outcomes of cancer immunity, autoimmunity, and chronic infections, the exact roles of these cells in various physiological and pathological processes remain elusive. Molecules that target and deplete PD-1+ cells would be instrumental in defining the roles unambiguously. Previously, an immunotoxin has been generated for the depletion of PD-1+ cells though its usage is impeded by its low production yield. Thus, a more practical molecular tool is desired to deplete PD-1+ cells and to examine functions of these cells. We designed and generated a novel anti-PD1 diphtheria immunotoxin, termed PD-1 DIT, targeting PD-1+ cells. PD-1 DIT is comprised of two single chain variable fragments (scFv) derived from an anti-PD-1 antibody, coupled with the catalytic and translocation domains of the diphtheria toxin. PD-1 DIT was produced using a yeast expression system that has been engineered to efficiently produce protein toxins. The yield of PD-1 DIT reached 1-2 mg/L culture, which is 10 times higher than the previously reported immunotoxin. Flow cytometry and confocal microscopy analyses confirmed that PD-1 DIT specifically binds to and enters PD-1+ cells. The binding avidities between PD-1 DIT and two PD-1+ cell lines are approximately 25 nM. Moreover, PD-1 DIT demonstrated potent cytotoxicity toward PD-1+ cells, with a half maximal effective concentration (EC50 ) value of 1 nM. In vivo experiments further showed that PD-1 DIT effectively depleted PD-1+ cells and enabled mice inoculated with PD-1+ tumor cells to survive throughout the study. Our findings using PD-1 DIT revealed the critical role of pancreatic PD-1+ T cells in the development of type-1 diabetes (T1D). Additionally, we observed that PD-1 DIT treatment ameliorated relapsing-remitting experimental autoimmune encephalomyelitis (RR-EAE), a mouse model of relapsing-remitting multiple sclerosis (RR-MS). Lastly, we did not observe significant hepatotoxicity in mice treated with PD-1 DIT, which had been reported for other immunotoxins derived from the diphtheria toxin. With its remarkable selective and potent cytotoxicity toward PD-1+ cells, coupled with its high production yield, PD-1 DIT emerges as a powerful biotechnological tool for elucidating the physiological roles of PD-1+ cells. Furthermore, the potential of PD-1 DIT to be developed into a novel therapeutic agent becomes evident.
Collapse
Affiliation(s)
- Tianxiao Zhang
- Department of Molecular PharmaceuticsUniversity of UtahSalt Lake CityUtahUSA
| | - Shuyun Dong
- Department of Molecular PharmaceuticsUniversity of UtahSalt Lake CityUtahUSA
| | - Yujia Zhai
- Department of Molecular PharmaceuticsUniversity of UtahSalt Lake CityUtahUSA
| | - Lauren Naatz
- Department of Molecular PharmaceuticsUniversity of UtahSalt Lake CityUtahUSA
| | - Zemin Zhou
- Department of PathologyUniversity of UtahSalt Lake CityUtahUSA
| | - Mingnan Chen
- Department of Molecular PharmaceuticsUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
5
|
Alkanlı SS, Dal Yöntem F, Yaşar M, Güven C, Kahraman MV, Kayaman Apohan N, Aktaş Z, Öncül MO, Ünlü A, Akçakaya H. Molecularly imprinted nanoparticles with recognition properties towards diphtheria toxin for ELISA applications. JOURNAL OF BIOMATERIALS SCIENCE, POLYMER EDITION 2022; 34:753-767. [PMID: 36357334 DOI: 10.1080/09205063.2022.2145866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Plastic antibodies can be used for in vitro neutralization of biomacromolecules with different fragments due to their potential in separation, purification, chemical sensor, catalysis and drug production studies. These polymer nanoparticles with binding affinity and selectivity comparable to natural antibodies were prepared using functional monomer synthesis and copolymerization of acrylic monomers via miniemulsion polymerization. As a result, the in vitro cytotoxic effect from diphtheria toxin was reduced by MIPs. In vitro imaging experiments of polymer nanoparticles (plastic antibodies) were performed to examine the interaction of diphtheria toxin with actin filaments, and MIPs inhibited diphtheria toxin damage on actin filaments. The enzyme-linked immunosorbent assay (ELISA) was performed with plastic antibodies labeled with biotin, and it was determined that plastic antibodies could also be used for diagnostic purposes. We report that molecularly imprinted polymers (MIPs), which are biocompatible polymer nanoparticles, can capture and reduce the effect of diphtheria toxic and its fragment A.
Collapse
Affiliation(s)
- Süleyman Serdar Alkanlı
- Department of Biophysics, Istanbul Faculty of Medicine, Istanbul University, 34093, Istanbul, Turkey
- Department of Biophysics, Institute of Health Sciences, Istanbul Faculty of Medicine, Istanbul University, 34093, Istanbul, Turkey
| | - Fulya Dal Yöntem
- Department of Biophysics, Koç University School of Medicine, Koç University, 34450, Sariyer, Istanbul, Turkey
- Koç University Research Center for Translational Medicine (KUTTAM), 34450, Sariyer, Istanbul, Turkey
| | - Merve Yaşar
- Department of Chemistry, Faculty of Art and Science, Marmara University, 34722, Göztepe, Istanbul, Turkey
| | - Celal Güven
- Department of Biophysics, Faculty of Medicine, Adiyaman University, 02040, Adiyaman, Turkey
| | - M. Vezir Kahraman
- Department of Chemistry, Faculty of Art and Science, Marmara University, 34722, Göztepe, Istanbul, Turkey
| | - Nilhan Kayaman Apohan
- Department of Chemistry, Faculty of Art and Science, Marmara University, 34722, Göztepe, Istanbul, Turkey
| | - Zerrin Aktaş
- Department of Microbiology & Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, 34104, Istanbul, Turkey
| | - Mustafa Oral Öncül
- Department of Infectious Diseases & Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, 34093, Istanbul, Turkey
| | - Ayhan Ünlü
- Department of Biophysics, Faculty of Medicine, Trakya University, 22020, Edirne, Turkey
| | - Handan Akçakaya
- Department of Biophysics, Istanbul Faculty of Medicine, Istanbul University, 34093, Istanbul, Turkey
| |
Collapse
|
6
|
Kulik M, Mori T, Sugita Y. Multi-Scale Flexible Fitting of Proteins to Cryo-EM Density Maps at Medium Resolution. Front Mol Biosci 2021; 8:631854. [PMID: 33842541 PMCID: PMC8025875 DOI: 10.3389/fmolb.2021.631854] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 01/26/2021] [Indexed: 11/13/2022] Open
Abstract
Structure determination using cryo-electron microscopy (cryo-EM) medium-resolution density maps is often facilitated by flexible fitting. Avoiding overfitting, adjusting force constants driving the structure to the density map, and emulating complex conformational transitions are major concerns in the fitting. To address them, we develop a new method based on a three-step multi-scale protocol. First, flexible fitting molecular dynamics (MD) simulations with coarse-grained structure-based force field and replica-exchange scheme between different force constants replicas are performed. Second, fitted Cα atom positions guide the all-atom structure in targeted MD. Finally, the all-atom flexible fitting refinement in implicit solvent adjusts the positions of the side chains in the density map. Final models obtained via the multi-scale protocol are significantly better resolved and more reliable in comparison with long all-atom flexible fitting simulations. The protocol is useful for multi-domain systems with intricate structural transitions as it preserves the secondary structure of single domains.
Collapse
Affiliation(s)
- Marta Kulik
- Theoretical Molecular Science Laboratory, RIKEN Cluster for Pioneering Research, Wako-shi, Japan
| | - Takaharu Mori
- Theoretical Molecular Science Laboratory, RIKEN Cluster for Pioneering Research, Wako-shi, Japan
| | - Yuji Sugita
- Theoretical Molecular Science Laboratory, RIKEN Cluster for Pioneering Research, Wako-shi, Japan.,RIKEN Center for Computational Science, Kobe, Japan.,RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
7
|
Rodnin MV, Kashipathy MM, Kyrychenko A, Battaile KP, Lovell S, Ladokhin AS. Structure of the Diphtheria Toxin at Acidic pH: Implications for the Conformational Switching of the Translocation Domain. Toxins (Basel) 2020; 12:toxins12110704. [PMID: 33171806 PMCID: PMC7695028 DOI: 10.3390/toxins12110704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/29/2020] [Accepted: 11/05/2020] [Indexed: 12/28/2022] Open
Abstract
Diphtheria toxin, an exotoxin secreted by Corynebacterium that causes disease in humans by inhibiting protein synthesis, enters the cell via receptor-mediated endocytosis. The subsequent endosomal acidification triggers a series of conformational changes, resulting in the refolding and membrane insertion of the translocation (T-)domain and ultimately leading to the translocation of the catalytic domain into the cytoplasm. Here, we use X-ray crystallography along with circular dichroism and fluorescence spectroscopy to gain insight into the mechanism of the early stages of pH-dependent conformational transition. For the first time, we present the high-resolution structure of the diphtheria toxin at a mildly acidic pH (5–6) and compare it to the structure at neutral pH (7). We demonstrate that neither catalytic nor receptor-binding domains change their structure upon this acidification, while the T-domain undergoes a conformational change that results in the unfolding of the TH2–3 helices. Surprisingly, the TH1 helix maintains its conformation in the crystal of the full-length toxin even at pH 5. This contrasts with the evidence from the new and previously published data, obtained by spectroscopic measurements and molecular dynamics computer simulations, which indicate the refolding of TH1 upon the acidification of the isolated T-domain. The overall results imply that the membrane interactions of the T-domain are critical in ensuring the proper conformational changes required for the preparation of the diphtheria toxin for the cellular entry.
Collapse
Affiliation(s)
- Mykola V. Rodnin
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (M.V.R.); (A.K.)
| | - Maithri M. Kashipathy
- Protein Structure Laboratory, Shankel Structural Biology Center, University of Kansas, Lawrence, KS 66047, USA; (M.M.K.); (S.L.)
| | - Alexander Kyrychenko
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (M.V.R.); (A.K.)
- Institute of Chemistry and School of Chemistry, V. N. Karazin Kharkiv National University, 61022 Kharkiv, Ukraine
| | - Kevin P. Battaile
- NYX beamline, New York Structural Biology Center, Upton, NY 11973, USA;
| | - Scott Lovell
- Protein Structure Laboratory, Shankel Structural Biology Center, University of Kansas, Lawrence, KS 66047, USA; (M.M.K.); (S.L.)
| | - Alexey S. Ladokhin
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (M.V.R.); (A.K.)
- Correspondence: ; Tel.: +1-913-588-0489; Fax: +1-913-588-7440
| |
Collapse
|
8
|
Westbrook JD, Soskind R, Hudson BP, Burley SK. Impact of the Protein Data Bank on antineoplastic approvals. Drug Discov Today 2020; 25:837-850. [PMID: 32068073 PMCID: PMC7305983 DOI: 10.1016/j.drudis.2020.02.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/08/2020] [Accepted: 02/07/2020] [Indexed: 12/14/2022]
Abstract
Open access to 3D structure information from the Protein Data Bank (PDB) facilitated discovery and development of >90% of the 79 new antineoplastic agents (54 small molecules, 25 biologics) with known molecular targets approved by the FDA 2010-2018. Analyses of PDB holdings, the scientific literature and related documents for each drug-target combination revealed that the impact of public-domain 3D structure data was broad and substantial, ranging from understanding target biology (∼95% of all targets) to identifying a given target as probably druggable (∼95% of all targets) to structure-guided lead optimization (>70% of all small-molecule drugs). In addition to aggregate impact assessments, illustrative case studies are presented for three protein kinase inhibitors, an allosteric enzyme inhibitor and seven advanced-stage melanoma therapeutics.
Collapse
Affiliation(s)
- John D Westbrook
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Rose Soskind
- Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Brian P Hudson
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Stephen K Burley
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA; Research Collaboratory for Structural Bioinformatics Protein Data Bank, San Diego Supercomputer Center, University of California, San Diego, La Jolla, CA 92093, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
9
|
Wenzel EV, Bosnak M, Tierney R, Schubert M, Brown J, Dübel S, Efstratiou A, Sesardic D, Stickings P, Hust M. Human antibodies neutralizing diphtheria toxin in vitro and in vivo. Sci Rep 2020; 10:571. [PMID: 31953428 PMCID: PMC6969050 DOI: 10.1038/s41598-019-57103-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/17/2019] [Indexed: 12/19/2022] Open
Abstract
Diphtheria is an infectious disease caused by Corynebacterium diphtheriae. The bacterium primarily infects the throat and upper airways and the produced diphtheria toxin (DT), which binds to the elongation factor 2 and blocks protein synthesis, can spread through the bloodstream and affect organs, such as the heart and kidneys. For more than 125 years, the therapy against diphtheria has been based on polyclonal horse sera directed against DT (diphtheria antitoxin; DAT). Animal sera have many disadvantages including serum sickness, batch-to-batch variation in quality and the use of animals for production. In this work, 400 human recombinant antibodies were generated against DT from two different phage display panning strategies using a human immune library. A panning in microtiter plates resulted in 22 unique in vitro neutralizing antibodies and a panning in solution combined with a functional neutralization screening resulted in 268 in vitro neutralizing antibodies. 61 unique antibodies were further characterized as scFv-Fc with 35 produced as fully human IgG1. The best in vitro neutralizing antibody showed an estimated relative potency of 454 IU/mg and minimal effective dose 50% (MED50%) of 3.0 pM at a constant amount of DT (4x minimal cytopathic dose) in the IgG format. The targeted domains of the 35 antibodies were analyzed by immunoblot and by epitope mapping using phage display. All three DT domains (enzymatic domain, translocation domain and receptor binding domain) are targets for neutralizing antibodies. When toxin neutralization assays were performed at higher toxin dose levels, the neutralizing capacity of individual antibodies was markedly reduced but this was largely compensated for by using two or more antibodies in combination, resulting in a potency of 79.4 IU/mg in the in vivo intradermal challenge assay. These recombinant antibody combinations are candidates for further clinical and regulatory development to replace equine DAT.
Collapse
Affiliation(s)
- Esther Veronika Wenzel
- Technische Universität Braunschweig, Institute for Biochemistry, Biotechnology and Bioinformatics, Department of Biotechnology, Braunschweig, Germany
| | - Margarita Bosnak
- Technische Universität Braunschweig, Institute for Biochemistry, Biotechnology and Bioinformatics, Department of Biotechnology, Braunschweig, Germany
| | - Robert Tierney
- National Institute for Biological Standards and Control (NIBSC), Division of Bacteriology, Potters Bar, United Kingdom
| | - Maren Schubert
- Technische Universität Braunschweig, Institute for Biochemistry, Biotechnology and Bioinformatics, Department of Biotechnology, Braunschweig, Germany
| | - Jeffrey Brown
- PETA International Science Consortium Ltd, London, United Kingdom
| | - Stefan Dübel
- Technische Universität Braunschweig, Institute for Biochemistry, Biotechnology and Bioinformatics, Department of Biotechnology, Braunschweig, Germany
| | - Androulla Efstratiou
- WHO Collaborating Centre for Diphtheria and Streptococcal Infections, London, UK
| | - Dorothea Sesardic
- National Institute for Biological Standards and Control (NIBSC), Division of Bacteriology, Potters Bar, United Kingdom
| | - Paul Stickings
- National Institute for Biological Standards and Control (NIBSC), Division of Bacteriology, Potters Bar, United Kingdom
| | - Michael Hust
- Technische Universität Braunschweig, Institute for Biochemistry, Biotechnology and Bioinformatics, Department of Biotechnology, Braunschweig, Germany.
| |
Collapse
|
10
|
Second-generation IL-2 receptor-targeted diphtheria fusion toxin exhibits antitumor activity and synergy with anti-PD-1 in melanoma. Proc Natl Acad Sci U S A 2019; 116:3100-3105. [PMID: 30718426 DOI: 10.1073/pnas.1815087116] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Denileukin diftitox (DAB-IL-2, Ontak) is a diphtheria-toxin-based fusion protein that depletes CD25-positive cells including regulatory T cells and has been approved for the treatment of persistent or recurrent cutaneous T cell lymphoma. However, the clinical use of denileukin diftitox was limited by vascular leak toxicity and production issues related to drug aggregation and purity. We found that a single amino acid substitution (V6A) in a motif associated with vascular leak induction yields a fully active, second-generation biologic, s-DAB-IL-2(V6A), which elicits 50-fold less human umbilical vein endothelial cell monolayer permeation and is 3.7-fold less lethal to mice by LD50 analysis than s-DAB-IL-2. Additionally, to overcome aggregation problems, we developed a production method for the fusion toxin using Corynebacterium diphtheriae that secretes fully folded, biologically active, monomeric s-DAB-IL-2 into the culture medium. Using the poorly immunogenic mouse B16F10 melanoma model, we initiated treatment 7 days after tumor challenge and observed that, while both s-DAB-IL-2(V6A) and s-DAB-IL-2 are inhibitors of tumor growth, the capacity to treat with higher doses of s-DAB-IL-2(V6A) could provide a superior activity window. In a sequential dual-therapy study in tumors that have progressed for 10 days, both s-DAB-IL-2(V6A) and s-DAB-IL-2 given before checkpoint inhibition with anti-programmed cell death-1 (anti-PD-1) antibodies inhibited tumor growth, while either drug given as monotherapy had less effect. s-DAB-IL-2(V6A), a fully monomeric protein with reduced vascular leak, is a second-generation diphtheria-toxin-based fusion protein with promise as a cancer immunotherapeutic both alone and in conjunction with PD-1 blockade.
Collapse
|
11
|
Structural and immunological characterization of E. coli derived recombinant CRM 197 protein used as carrier in conjugate vaccines. Biosci Rep 2018; 38:BSR20180238. [PMID: 29875175 PMCID: PMC6153374 DOI: 10.1042/bsr20180238] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 05/21/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022] Open
Abstract
It is established that the immunogenicity of polysaccharides is enhanced by coupling them to carrier proteins. Cross reacting material (CRM197), a nontoxic variant of diphtheria toxin (DT) is widely used carrier protein for polysaccharide conjugate vaccines. Conventionally, CRM197 is isolated by fermentation of Corynebacterium diphtheriae C7 (β197) cultures, which often suffers from low yield. Recently, several recombinant approaches have been reported with robust processes and higher yields, which will improve the affordability of CRM197-based vaccines. Vaccine manufacturers require detailed analytical information to ensure that the CRM197 meets quality standards and regulatory requirements. In the present manuscript we have described detailed structural characteristics of Escherichia coli based recombinant CRM197 (rCRM197) carrier protein. The crystal structure of the E. coli based rCRM197 was found to be identical with the reported crystal structure of the C7 CRM197 produced in C. diphtheriae C7 strain (Protein Data Bank (PDB) ID: 4EA0). The crystal structure of rCRM197 was determined at 2.3 Å resolution and structure was submitted to the PDB with accession number ID 5I82. This is the first report of a crystal structure of E. coli derived recombinant CRM197 carrier protein. Furthermore, the rCRM197 was conjugated to Vi polysaccharide to generate Typhoid conjugate vaccine (Vi-rCRM197) and its immunogenicity was evaluated in Balb/C Mice. The Vi-rCRM197 conjugate vaccine was found to generate strong primary α-Vi antibody response and also showed a booster response after subsequent vaccination in mice. Overall data suggest that E. coli based recombinant CRM197 exhibits structural and immunological similarity with the C7 CRM197 and can be used as a carrier protein in conjugate vaccine development.
Collapse
|
12
|
Refining Protein Penetration into the Lipid Bilayer Using Fluorescence Quenching and Molecular Dynamics Simulations: The Case of Diphtheria Toxin Translocation Domain. J Membr Biol 2018; 251:379-391. [PMID: 29550876 DOI: 10.1007/s00232-018-0030-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 03/11/2018] [Indexed: 12/23/2022]
Abstract
Dynamic disorder of the lipid bilayer presents a challenge for establishing structure-function relationships in membranous systems. The resulting structural heterogeneity is especially evident for peripheral and spontaneously inserting membrane proteins, which are not constrained by the well-defined transmembrane topology and exert their action in the context of intimate interaction with lipids. Here, we propose a concerted approach combining depth-dependent fluorescence quenching with Molecular Dynamics simulation to decipher dynamic interactions of membrane proteins with the lipid bilayers. We apply this approach to characterize membrane-mediated action of the diphtheria toxin translocation domain. First, we use a combination of the steady-state and time-resolved fluorescence spectroscopy to characterize bilayer penetration of the NBD probe selectively attached to different sites of the protein into membranes containing lipid-attached nitroxyl quenching groups. The constructed quenching profiles are analyzed with the Distribution Analysis methodology allowing for accurate determination of transverse distribution of the probe. The results obtained for 12 NBD-labeled single-Cys mutants are consistent with the so-called Open-Channel topology model. The experimentally determined quenching profiles for labeling sites corresponding to L350, N373, and P378 were used as initial constraints for positioning TH8-9 hairpin into the lipid bilayer for Molecular Dynamics simulation. Finally, we used alchemical free energy calculations to characterize protonation of E362 in soluble translocation domain and membrane-inserted conformation of its TH8-9 fragment. Our results indicate that membrane partitioning of the neutral E362 is more favorable energetically (by ~ 6 kcal/mol), but causes stronger perturbation of the bilayer, than the charged E362.
Collapse
|
13
|
Wittchen M, Busche T, Gaspar AH, Lee JH, Ton-That H, Kalinowski J, Tauch A. Transcriptome sequencing of the human pathogen Corynebacterium diphtheriae NCTC 13129 provides detailed insights into its transcriptional landscape and into DtxR-mediated transcriptional regulation. BMC Genomics 2018; 19:82. [PMID: 29370758 PMCID: PMC5784534 DOI: 10.1186/s12864-018-4481-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 01/16/2018] [Indexed: 12/27/2022] Open
Abstract
Background The human pathogen Corynebacterium diphtheriae is the causative agent of diphtheria. In the 1990s a large diphtheria outbreak in Eastern Europe was caused by the strain C. diphtheriae NCTC 13129. Although the genome was sequenced more than a decade ago, not much is known about its transcriptome. Our aim was to use transcriptome sequencing (RNA-Seq) to close this knowledge gap and gain insights into the transcriptional landscape of a C. diphtheriae tox+ strain. Results We applied two different RNA-Seq techniques, one to retrieve 5′-ends of primary transcripts and the other to characterize the whole transcriptional landscape in order to gain insights into various features of the C. diphtheriae NCTC 13129 transcriptome. By examining the data we identified 1656 transcription start sites (TSS), of which 1202 were assigned to genes and 454 to putative novel transcripts. By using the TSS data promoter regions recognized by the housekeeping sigma factor σA and its motifs were analyzed in detail, revealing a well conserved −10 but an only weakly conserved −35 motif, respectively. Furthermore, with the TSS data 5’-UTR lengths were explored. The observed 5’-UTRs range from zero length (leaderless transcripts), which make up 20% of all genes, up to over 450 nt long leaders, which may harbor regulatory functions. The C. diphtheriae transcriptome consists of 471 operons which are further divided into 167 sub-operon structures. In a differential expression analysis approach, we discovered that genetic disruption of the iron-sensing transcription regulator DtxR, which controls expression of diphtheria toxin (DT), causes a strong influence on general gene expression. Nearly 15% of the genome is differentially transcribed, indicating that DtxR might have other regulatory functions in addition to regulation of iron metabolism and DT. Furthermore, our findings shed light on the transcriptional landscape of the DT encoding gene tox and present evidence for two tox antisense RNAs, which point to a new way of transcriptional regulation of toxin production. Conclusions This study presents extensive insights into the transcriptome of C. diphtheriae and provides a basis for future studies regarding gene characterization, transcriptional regulatory networks, and regulation of the tox gene in particular. Electronic supplementary material The online version of this article (10.1186/s12864-018-4481-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manuel Wittchen
- Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Tobias Busche
- Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany.,Institute for Biology-Microbiology, Freie Universität Berlin, D-14195, Berlin, Germany
| | - Andrew H Gaspar
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, USA
| | - Ju Huck Lee
- Department of Microbiology & Molecular Genetics, University of Texas McGovern Medical School at Houston, Houston, USA.,Present address: Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeollabuk-do, 56212, Republic of Korea
| | - Hung Ton-That
- Department of Microbiology & Molecular Genetics, University of Texas McGovern Medical School at Houston, Houston, USA
| | - Jörn Kalinowski
- Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany.
| | - Andreas Tauch
- Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| |
Collapse
|
14
|
Cellular Entry of the Diphtheria Toxin Does Not Require the Formation of the Open-Channel State by Its Translocation Domain. Toxins (Basel) 2017; 9:toxins9100299. [PMID: 28937631 PMCID: PMC5666346 DOI: 10.3390/toxins9100299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/20/2017] [Accepted: 09/20/2017] [Indexed: 12/30/2022] Open
Abstract
Cellular entry of diphtheria toxin is a multistage process involving receptor targeting, endocytosis, and translocation of the catalytic domain across the endosomal membrane into the cytosol. The latter is ensured by the translocation (T) domain of the toxin, capable of undergoing conformational refolding and membrane insertion in response to the acidification of the endosomal environment. While numerous now classical studies have demonstrated the formation of an ion-conducting conformation-the Open-Channel State (OCS)-as the final step of the refolding pathway, it remains unclear whether this channel constitutes an in vivo translocation pathway or is a byproduct of the translocation. To address this question, we measure functional activity of known OCS-blocking mutants with H-to-Q replacements of C-terminal histidines of the T-domain. We also test the ability of these mutants to translocate their own N-terminus across lipid bilayers of model vesicles. The results of both experiments indicate that translocation activity does not correlate with previously published OCS activity. Finally, we determined the topology of TH5 helix in membrane-inserted T-domain using W281 fluorescence and its depth-dependent quenching by brominated lipids. Our results indicate that while TH5 becomes a transbilayer helix in a wild-type protein, it fails to insert in the case of the OCS-blocking mutant H322Q. We conclude that the formation of the OCS is not necessary for the functional translocation by the T-domain, at least in the histidine-replacement mutants, suggesting that the OCS is unlikely to constitute a translocation pathway for the cellular entry of diphtheria toxin in vivo.
Collapse
|
15
|
Alsarraf H, Dedic E, Bjerrum MJ, Østergaard O, Kristensen MP, Petersen JW, Jørgensen R. Biophysical comparison of diphtheria and tetanus toxins with the formaldehyde-detoxified toxoids, the main components of diphtheria and tetanus vaccines. Virulence 2017; 8:1880-1889. [PMID: 28430538 DOI: 10.1080/21505594.2017.1321726] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Husam Alsarraf
- a Department of Microbiology and Infection Control , Statens Serum Institut , Copenhagen S. , Denmark
| | - Emil Dedic
- a Department of Microbiology and Infection Control , Statens Serum Institut , Copenhagen S. , Denmark
| | - Morten J Bjerrum
- b Department of Chemistry , University of Copenhagen , Copenhagen Ø. , Denmark
| | - Ole Østergaard
- c Department of Autoimmunology , Statens Serum Institut , Copenhagen S. , Denmark
| | - Max Per Kristensen
- d Vaccine Development Department , Statens Serum Institut , Copenhagen S. , Denmark
| | - Jesper W Petersen
- e Bacterial Vaccine Department , Statens Serum Institut , Copenhagen S. , Denmark
| | - René Jørgensen
- a Department of Microbiology and Infection Control , Statens Serum Institut , Copenhagen S. , Denmark
| |
Collapse
|
16
|
Miyashita O, Kobayashi C, Mori T, Sugita Y, Tama F. Flexible fitting to cryo-EM density map using ensemble molecular dynamics simulations. J Comput Chem 2017; 38:1447-1461. [PMID: 28370077 DOI: 10.1002/jcc.24785] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 01/26/2017] [Accepted: 02/22/2017] [Indexed: 12/25/2022]
Abstract
Flexible fitting is a computational algorithm to derive a new conformational model that conforms to low-resolution experimental data by transforming a known structure. A common application is against data from cryo-electron microscopy to obtain conformational models in new functional states. The conventional flexible fitting algorithms cannot derive correct structures in some cases due to the complexity of conformational transitions. In this study, we show the importance of conformational ensemble in the refinement process by performing multiple fittings trials using a variety of different force constants. Application to simulated maps of Ca2+ ATPase and diphtheria toxin as well as experimental data of release factor 2 revealed that for these systems, multiple conformations with similar agreement with the density map exist and a large number of fitting trials are necessary to generate good models. Clustering analysis can be an effective approach to avoid over-fitting models. In addition, we show that an automatic adjustment of the biasing force constants during the fitting process, implemented as replica-exchange scheme, can improve the success rate. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Osamu Miyashita
- Advanced Institute for Computational Science, RIKEN, 6-7-1, Minatojima-minami-machi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Chigusa Kobayashi
- Advanced Institute for Computational Science, RIKEN, 6-7-1, Minatojima-minami-machi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Takaharu Mori
- Theoretical Molecular Science Laboratory, RIKEN, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan.,iTHES, RIKEN, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Yuji Sugita
- Advanced Institute for Computational Science, RIKEN, 6-7-1, Minatojima-minami-machi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.,Theoretical Molecular Science Laboratory, RIKEN, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan.,iTHES, RIKEN, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan.,Quantitative Biology Center, RIKEN, 6-7-1, Minatojima-minami-machi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Florence Tama
- Advanced Institute for Computational Science, RIKEN, 6-7-1, Minatojima-minami-machi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.,Department of Physics and ITbM, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi, 464-8602, Japan
| |
Collapse
|
17
|
Mascarenhas NM, Gosavi S. Understanding protein domain-swapping using structure-based models of protein folding. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 128:113-120. [PMID: 27867057 PMCID: PMC7127520 DOI: 10.1016/j.pbiomolbio.2016.09.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 09/05/2016] [Accepted: 09/26/2016] [Indexed: 02/05/2023]
Abstract
In domain-swapping, two or more identical protein monomers exchange structural elements and fold into dimers or multimers whose units are structurally similar to the original monomer. Domain-swapping is of biotechnological interest because inhibiting domain-swapping can reduce disease-causing fibrillar protein aggregation. To achieve such inhibition, it is important to understand both the energetics that stabilize the domain-swapped structure and the protein dynamics that enable the swapping. Structure-based models (SBMs) encode the folded structure of the protein in their potential energy functions. SBMs have been successfully used to understand diverse aspects of monomer folding. Symmetrized SBMs model interactions between two identical protein chains using only intra-monomer interactions. Molecular dynamics simulations of such symmetrized SBMs have been used to correctly predict the domain-swapped structure and to understand the mechanism of domain-swapping. Here, we review such models and illustrate that monomer topology determines key aspects of domain-swapping. However, in some proteins, specifics of local energetic interactions modulate domain-swapping and these need to be added to the symmetrized SBMs. We then summarize some general principles of the mechanism of domain-swapping that emerge from the symmetrized SBM simulations. Finally, using our own results, we explore how symmetrized SBMs could be used to design domain-swapping in proteins.
Collapse
Affiliation(s)
- Nahren Manuel Mascarenhas
- Simons Centre for the Study of Living Machines, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Shachi Gosavi
- Simons Centre for the Study of Living Machines, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India.
| |
Collapse
|
18
|
Assar Z, Nossoni Z, Wang W, Santos EM, Kramer K, McCornack C, Vasileiou C, Borhan B, Geiger JH. Domain-Swapped Dimers of Intracellular Lipid-Binding Proteins: Evidence for Ordered Folding Intermediates. Structure 2016; 24:1590-8. [PMID: 27524203 PMCID: PMC5330279 DOI: 10.1016/j.str.2016.05.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/13/2016] [Accepted: 05/16/2016] [Indexed: 11/29/2022]
Abstract
Human Cellular Retinol Binding Protein II (hCRBPII), a member of the intracellular lipid-binding protein family, is a monomeric protein responsible for the intracellular transport of retinol and retinal. Herein we report that hCRBPII forms an extensive domain-swapped dimer during bacterial expression. The domain-swapped region encompasses almost half of the protein. The dimer represents a novel structural architecture with the mouths of the two binding cavities facing each other, producing a new binding cavity that spans the length of the protein complex. Although wild-type hCRBPII forms the dimer, the propensity for dimerization can be substantially increased via mutation at Tyr60. The monomeric form of the wild-type protein represents the thermodynamically more stable species, making the domain-swapped dimer a kinetically trapped entity. Hypothetically, the wild-type protein has evolved to minimize dimerization of the folding intermediate through a critical hydrogen bond (Tyr60-Glu72) that disfavors the dimeric form.
Collapse
Affiliation(s)
- Zahra Assar
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Zahra Nossoni
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Wenjing Wang
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Elizabeth M Santos
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Kevin Kramer
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Colin McCornack
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Chrysoula Vasileiou
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - Babak Borhan
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA.
| | - James H Geiger
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
19
|
Crystallographic studies on protein misfolding: Domain swapping and amyloid formation in the SH3 domain. Arch Biochem Biophys 2016; 602:116-126. [PMID: 26924596 DOI: 10.1016/j.abb.2016.02.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 02/19/2016] [Accepted: 02/23/2016] [Indexed: 12/18/2022]
Abstract
Oligomerization by 3D domain swapping is found in a variety of proteins of diverse size, fold and function. In the early 1960s this phenomenon was postulated for the oligomers of ribonuclease A, but it was not until the 1990s that X-ray diffraction provided the first experimental evidence of this special manner of oligomerization. Nowadays, structural information has allowed the identification of these swapped oligomers in over one hundred proteins. Although the functional relevance of this phenomenon is not clear, this alternative folding of protomers into intertwined oligomers has been related to amyloid formation. Studies on proteins that develop 3D domain swapping might provide some clues on the early stages of amyloid formation. The SH3 domain is a small modular domain that has been used as a model to study the basis of protein folding. Among SH3 domains, the c-Src-SH3 domain emerges as a helpful model to study 3D domain swapping and amyloid formation.
Collapse
|
20
|
Topography of the TH5 Segment in the Diphtheria Toxin T-Domain Channel. J Membr Biol 2015; 249:181-96. [PMID: 26645703 DOI: 10.1007/s00232-015-9859-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/18/2015] [Indexed: 10/22/2022]
Abstract
The translocation domain (T-domain) of diphtheria toxin contains 10 α helices in the aqueous crystal structure. Upon exposure to a planar lipid bilayer under acidic conditions, it inserts to form a channel and transport the attached amino-terminal catalytic domain across the membrane. The TH5, TH8, and TH9 helices form transmembrane segments in the open-channel state, with TH1-TH4 translocated across the membrane. The TH6-TH7 segment also inserts to form a constriction that occupies only a small portion of the total channel length. Here, we have examined the TH5 segment in more detail, using the substituted-cysteine accessibility method. We constructed a series of 23 mutant T-domains with single cysteine residues at positions in and near TH5, monitored their channel formation in planar lipid bilayers, and probed for an effect of thiol-specific reagents added to the solutions on either side of the membrane. For 15 of the mutants, the reagent caused a decrease in single-channel conductance, indicating that the introduced cysteine residue was exposed within the channel lumen. We also found that reaction caused large changes in ionic selectivity for some mutant channels. We determined whether reaction occurred in the open state or in the brief flicker-closed state of the channel. Finally, we compared the reaction rates from either side of the membrane. Our experiments are consistent with the hypotheses that the TH5 helix has a transmembrane orientation and remains helical in the open-channel state; they also indicate that the middle of the helix is aligned with the constriction in the channel.
Collapse
|
21
|
Seyler SL, Kumar A, Thorpe MF, Beckstein O. Path Similarity Analysis: A Method for Quantifying Macromolecular Pathways. PLoS Comput Biol 2015; 11:e1004568. [PMID: 26488417 PMCID: PMC4619321 DOI: 10.1371/journal.pcbi.1004568] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 09/23/2015] [Indexed: 01/03/2023] Open
Abstract
Diverse classes of proteins function through large-scale conformational changes and various sophisticated computational algorithms have been proposed to enhance sampling of these macromolecular transition paths. Because such paths are curves in a high-dimensional space, it has been difficult to quantitatively compare multiple paths, a necessary prerequisite to, for instance, assess the quality of different algorithms. We introduce a method named Path Similarity Analysis (PSA) that enables us to quantify the similarity between two arbitrary paths and extract the atomic-scale determinants responsible for their differences. PSA utilizes the full information available in 3N-dimensional configuration space trajectories by employing the Hausdorff or Fréchet metrics (adopted from computational geometry) to quantify the degree of similarity between piecewise-linear curves. It thus completely avoids relying on projections into low dimensional spaces, as used in traditional approaches. To elucidate the principles of PSA, we quantified the effect of path roughness induced by thermal fluctuations using a toy model system. Using, as an example, the closed-to-open transitions of the enzyme adenylate kinase (AdK) in its substrate-free form, we compared a range of protein transition path-generating algorithms. Molecular dynamics-based dynamic importance sampling (DIMS) MD and targeted MD (TMD) and the purely geometric FRODA (Framework Rigidity Optimized Dynamics Algorithm) were tested along with seven other methods publicly available on servers, including several based on the popular elastic network model (ENM). PSA with clustering revealed that paths produced by a given method are more similar to each other than to those from another method and, for instance, that the ENM-based methods produced relatively similar paths. PSA was applied to ensembles of DIMS MD and FRODA trajectories of the conformational transition of diphtheria toxin, a particularly challenging example. For the AdK transition, the new concept of a Hausdorff-pair map enabled us to extract the molecular structural determinants responsible for differences in pathways, namely a set of conserved salt bridges whose charge-charge interactions are fully modelled in DIMS MD but not in FRODA. PSA has the potential to enhance our understanding of transition path sampling methods, validate them, and to provide a new approach to analyzing conformational transitions. Many proteins are nanomachines that perform mechanical or chemical work by changing their three-dimensional shape and cycle between multiple conformational states. Computer simulations of such conformational transitions provide mechanistic insights into protein function but such simulations have been challenging. In particular, it is not clear how to quantitatively compare current simulation methods or to assess their accuracy. To that end, we present a general and flexible computational framework for quantifying transition paths—by measuring mutual geometric similarity—that, compared with existing approaches, requires minimal a-priori assumptions and can take advantage of full atomic detail alongside heuristic information derived from intuition. Using our Path Similarity Analysis (PSA) framework in parallel with several existing quantitative approaches, we examine transitions generated for a toy model of a transition and two biological systems, the enzyme adenylate kinase and diphtheria toxin. Our results show that PSA enables the quantitative comparison of different path sampling methods and aids the identification of potentially important atomistic motions by exploiting geometric information in transition paths. The method has the potential to enhance our understanding of transition path sampling methods, validate them, and to provide a new approach to analyzing macromolecular conformational transitions.
Collapse
Affiliation(s)
- Sean L. Seyler
- Department of Physics and Center for Biological Physics, Arizona State University, Tempe, Arizona, United States of America
| | - Avishek Kumar
- Department of Physics and Center for Biological Physics, Arizona State University, Tempe, Arizona, United States of America
| | - M. F. Thorpe
- Department of Physics and Center for Biological Physics, Arizona State University, Tempe, Arizona, United States of America
- Rudolf Peierls Centre for Theoretical Physics, University of Oxford, Oxford, United Kingdom
| | - Oliver Beckstein
- Department of Physics and Center for Biological Physics, Arizona State University, Tempe, Arizona, United States of America
- * E-mail:
| |
Collapse
|
22
|
Labyntsev AI, Korotkevich NV, Manoĭlov KI, Kaberniuk AA, Kolibo DV, Komisarenko SV. [Recombinant fluorescent models for studying of diphtheria toxin]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2015; 40:433-42. [PMID: 25898753 DOI: 10.1134/s1068162014040086] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Diphtheria toxin is the main pathogenicity factor of causative agent of diphtheria Corynebacterium diphtheriae. Due to the small molecule size, it is of considerable interestfor the development of synthetic protein molecules with transporting function, e.g. immunotoxins. Expression and characterization of nontoxic recombinant fluorescent derivates of diphtheria toxin and its nontoxic mutant CRM 197 were described in this article. Obtained proteins may be applied in studies of receptor-binding and transporting functions of the toxin in cells, for determination of toxin receptor proHB-EGF expression level, immunization and antibody generation against the toxin and in development of diagnostic test-systems, detection of diphtheria toxin and antitoxic antibodies.
Collapse
|
23
|
Kienker PK, Wu Z, Finkelstein A. Mapping the membrane topography of the TH6-TH7 segment of the diphtheria toxin T-domain channel. ACTA ACUST UNITED AC 2015; 145:107-25. [PMID: 25582482 PMCID: PMC4306713 DOI: 10.1085/jgp.201411326] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cysteine substitution accessibility analysis suggests that the TH6–TH7 segment forms a constriction in the diphtheria toxin T-domain channel. Low pH triggers the translocation domain of diphtheria toxin (T-domain), which contains 10 α helices, to insert into a planar lipid bilayer membrane, form a transmembrane channel, and translocate the attached catalytic domain across the membrane. Three T-domain helices, corresponding to TH5, TH8, and TH9 in the aqueous crystal structure, form transmembrane segments in the open-channel state; the amino-terminal region, TH1–TH4, translocates across the membrane to the trans side. Residues near either end of the TH6–TH7 segment are not translocated, remaining on the cis side of the membrane; because the intervening 25-residue sequence is too short to form a transmembrane α-helical hairpin, it was concluded that the TH6–TH7 segment resides at the cis interface. Now we have examined this segment further, using the substituted-cysteine accessibility method. We constructed a series of 18 mutant T-domains with single cysteine residues at positions in TH6–TH7, monitored their channel formation in planar lipid bilayers, and probed for an effect of thiol-specific reagents on the channel conductance. For 10 of the mutants, the reagent caused a change in the single-channel conductance, indicating that the introduced cysteine residue was exposed within the channel lumen. For several of these mutants, we verified that the reactions occurred primarily in the open state, rather than in the flicker-closed state. We also established that blocking of the channel by an amino-terminal hexahistidine tag could protect mutants from reaction. Finally, we compared the reaction rates of reagent added to the cis and trans sides to quantify the residue’s accessibility from either side. This analysis revealed abrupt changes in cis- versus trans-side accessibility, suggesting that the TH6–TH7 segment forms a constriction that occupies a small portion of the total channel length. We also determined that this constriction is located near the middle of the TH8 helix.
Collapse
Affiliation(s)
- Paul K Kienker
- Department of Physiology and Biophysics, and Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Zhengyan Wu
- Department of Physiology and Biophysics, and Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Alan Finkelstein
- Department of Physiology and Biophysics, and Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461 Department of Physiology and Biophysics, and Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
24
|
The influence of flanking secondary structures on amino Acid content and typical lengths of 3/10 helices. INTERNATIONAL JOURNAL OF PROTEOMICS 2014; 2014:360230. [PMID: 25371821 PMCID: PMC4211214 DOI: 10.1155/2014/360230] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/19/2014] [Accepted: 09/27/2014] [Indexed: 11/25/2022]
Abstract
We used 3D structures of a highly redundant set of bacterial proteins encoded by genes of high, average, and low GC-content. Four types of connecting bridges—regions situated between any of two major elements of secondary structure (alpha helices and beta strands)—containing a pure random coil were compared with connecting bridges containing 3/10 helices. We included discovered trends in the original “VVTAK Connecting Bridges” algorithm, which is able to predict more probable conformation for a given connecting bridge. The highest number of significant differences in amino acid usage was found between 3/10 helices containing bridges connecting two beta strands (they have increased Phe, Tyr, Met, Ile, Leu, Val, and His usages but decreased usages of Asp, Asn, Gly, and Pro) and those without 3/10 helices. The typical (most common) length of 3/10 helices situated between two beta strands and between beta strand and alpha helix is equal to 5 amino acid residues. The preferred length of 3/10 helices situated between alpha helix and beta strand is equal to 3 residues. For 3/10 helices situated between two alpha helices, both lengths (3 and 5 amino acid residues) are typical.
Collapse
|
25
|
Peter B, Polyansky AA, Fanucchi S, Dirr HW. A Lys-Trp cation-π interaction mediates the dimerization and function of the chloride intracellular channel protein 1 transmembrane domain. Biochemistry 2013; 53:57-67. [PMID: 24328417 DOI: 10.1021/bi401433f] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chloride intracellular channel protein 1 (CLIC1) is a dual-state protein that can exist either as a soluble monomer or in an integral membrane form. The oligomerization of the transmembrane domain (TMD) remains speculative despite it being implicated in pore formation. The extent to which electrostatic and van der Waals interactions drive folding and association of the dimorphic TMD is unknown and is complicated by the requirement of interactions favorable in both aqueous and membrane environments. Here we report a putative Lys37-Trp35 cation-π interaction and show that it stabilizes the dimeric form of the CLIC1 TMD in membranes. A synthetic 30-mer peptide comprising a K37M TMD mutant was examined in 2,2,2-trifluoroethanol, sodium dodecyl sulfate micelles, and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine liposomes using far-ultraviolet (UV) circular dichroism, fluorescence, and UV absorbance spectroscopy. Our data suggest that Lys37 is not implicated in the folding, stability, or membrane insertion of the TMD peptide. However, removal of this residue impairs the formation of dimers and higher-order oligomers. This is accompanied by a 30-fold loss of chloride influx activity, suggesting that dimerization modulates the rate of chloride conductance. We propose that, within membranes, individual TMD helices associate via a Lys37-mediated cation-π interaction to form active dimers. The latter findings are also supported by results of modeling a putative TMD dimer conformation in which Lys37 and Trp35 form cation-π pairs at the dimer interface. Dimeric helix bundles may then associate to form fully active ion channels. Thus, within a membrane-like environment, aromatic interactions involving a polar lysine side chain provide a thermodynamic driving force for helix-helix association.
Collapse
Affiliation(s)
- Bradley Peter
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand , Johannesburg 2050, South Africa
| | | | | | | |
Collapse
|
26
|
Farelli JD, Gumbart JC, Akey IV, Hempstead A, Amyot W, Head JF, McKnight CJ, Isberg RR, Akey CW. IcmQ in the Type 4b secretion system contains an NAD+ binding domain. Structure 2013; 21:1361-73. [PMID: 23850453 DOI: 10.1016/j.str.2013.05.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Revised: 05/06/2013] [Accepted: 05/26/2013] [Indexed: 12/23/2022]
Abstract
A Type 4b secretion system (T4bSS) is required for Legionella growth in alveolar macrophages. IcmQ associates with IcmR, binds to membranes, and has a critical role in the T4bSS. We have now solved a crystal structure of IcmR-IcmQ to further our understanding of this complex. This structure revealed an amphipathic four-helix bundle, formed by IcmR and the N-terminal domain of IcmQ, which is linked to a novel C-terminal domain of IcmQ (Qc) by a linker helix. The Qc domain has structural homology with ADP ribosyltransferase domains in certain bacterial toxins and binds NAD(+) with a dissociation constant in the physiological range. Structural homology and molecular dynamics were used to identify an extended NAD(+) binding site on Qc, and the resulting model was tested by mutagenesis and binding assays. Based on the data, we suggest that IcmR-IcmQ binds to membranes, where it may interact with, or perhaps modify, a protein in the T4bSS when NAD(+) is bound.
Collapse
Affiliation(s)
- Jeremiah D Farelli
- Department of Physiology and Biophysics, Boston University School of Medicine, 700 Albany Street, Boston, MA 02118-2526, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Peter B, Ngubane NCML, Fanucchi S, Dirr HW. Membrane Mimetics Induce Helix Formation and Oligomerization of the Chloride Intracellular Channel Protein 1 Transmembrane Domain. Biochemistry 2013; 52:2739-49. [DOI: 10.1021/bi4002776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Bradley Peter
- Protein Structure-Function Research
Unit, School of
Molecular and Cell Biology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Nomxolisi Chloë Mina-Liz Ngubane
- Protein Structure-Function Research
Unit, School of
Molecular and Cell Biology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Sylvia Fanucchi
- Protein Structure-Function Research
Unit, School of
Molecular and Cell Biology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Heini W. Dirr
- Protein Structure-Function Research
Unit, School of
Molecular and Cell Biology, University of the Witwatersrand, Johannesburg 2050, South Africa
| |
Collapse
|
28
|
Mathiharan YK, Pappachan A, Savithri HS, Murthy MRN. Dramatic structural changes resulting from the loss of a crucial hydrogen bond in the hinge region involved in C-terminal helix swapping in SurE: a survival protein from Salmonella typhimurium. PLoS One 2013; 8:e55978. [PMID: 23409101 PMCID: PMC3567009 DOI: 10.1371/journal.pone.0055978] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 01/04/2013] [Indexed: 11/30/2022] Open
Abstract
Domain swapping is an interesting feature of some oligomeric proteins in which each protomer of the oligomer provides an identical surface for exclusive interaction with a segment or domain belonging to another protomer. Here we report results of mutagenesis experiments on the structure of C-terminal helix swapped dimer of a stationary phase survival protein from Salmonella typhimurium (StSurE). Wild type StSurE is a dimer in which a large helical segment at the C-terminus and a tetramerization loop comprising two β strands are swapped between the protomers. Key residues in StSurE that might promote C-terminal helix swapping were identified by sequence and structural comparisons. Three mutants in which the helix swapping is likely to be avoided were constructed and expressed in E. coli. Three-dimensional X-ray crystal structures of the mutants H234A and D230A/H234A could be determined at 2.1 Å and 2.35 Å resolutions, respectively. Contrary to expectations, helix swapping was mostly retained in both the mutants. The loss of the crucial D230 OD2– H234 NE2 hydrogen bond (2.89 Å in the wild type structure) in the hinge region was compensated by new inter and intra-chain interactions. However, the two fold molecular symmetry was lost and there were large conformational changes throughout the polypeptide. In spite of these changes, the dimeric structure and an approximate tetrameric organization were retained, probably due to the interactions involving the tetramerization loop. Mutants were mostly functionally inactive, highlighting the importance of precise inter-subunit interactions for the symmetry and function of StSurE.
Collapse
Affiliation(s)
| | - Anju Pappachan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - H. S. Savithri
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Mathur R. N. Murthy
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
- * E-mail:
| |
Collapse
|
29
|
Matar AJ, Pathiraja V, Wang Z, Duran-Struuck R, Gusha A, Crepeau R, Tasaki M, Sachs DH, Huang CA. Effect of pre-existing anti-diphtheria toxin antibodies on T cell depletion levels following diphtheria toxin-based recombinant anti-monkey CD3 immunotoxin treatment. Transpl Immunol 2012; 27:52-4. [PMID: 22676970 DOI: 10.1016/j.trim.2012.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 05/17/2012] [Accepted: 05/21/2012] [Indexed: 10/28/2022]
Abstract
Diphtheria toxin (DT)-based anti-CD3 immunotoxins have clinical relevance in numerous applications including autoimmune disease therapies and organ transplantation tolerance protocols. Pre-existing anti-DT antibodies acquired either by vaccination against diphtheria toxin or infections with C. diphtheriae may interfere or inhibit the function of these anti-CD3 immunotoxins. Previously, a full-length anti-rhesus monkey CD3 immunotoxin, FN18-CRM9, was shown to be less effective at depleting circulating T cells in animals with pre-existing anti-DT antibody titers than in animals without antibodies, and subsequent doses were ineffective. In this study, the T cell depletion function of a truncated DT based recombinant anti-monkey CD3 immunotoxin, A-dmDT390-scfbDb (C207), as part of a reduced intensity conditioning regimen prior to hematopoietic cell transplantation, was compared between two groups of monkeys: those with and without pre-existing anti-diphtheria titers. T cell depletion was comparable in both groups of monkeys, and therefore appeared to be unaffected by the presence of moderate levels of pre-existing anti-diphtheria antibodies.
Collapse
Affiliation(s)
- Abraham J Matar
- Transplantation Biology Research Center, Massachusetts General Hospital, Boston, MA, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Arai R, Kobayashi N, Kimura A, Sato T, Matsuo K, Wang AF, Platt JM, Bradley LH, Hecht MH. Domain-Swapped Dimeric Structure of a Stable and Functional De Novo Four-Helix Bundle Protein, WA20. J Phys Chem B 2012; 116:6789-97. [DOI: 10.1021/jp212438h] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ryoichi Arai
- International
Young Researchers
Empowerment Center, Shinshu University,
Ueda, Nagano 386-8567, Japan
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544,
United States
- Department of Applied Biology, Faculty of Textile Science & Technology, Shinshu University, Ueda, Nagano 386-8567, Japan
| | - Naoya Kobayashi
- Department of Applied Biology, Faculty of Textile Science & Technology, Shinshu University, Ueda, Nagano 386-8567, Japan
| | - Akiho Kimura
- Department of Applied Biology, Faculty of Textile Science & Technology, Shinshu University, Ueda, Nagano 386-8567, Japan
| | - Takaaki Sato
- International
Young Researchers
Empowerment Center, Shinshu University,
Ueda, Nagano 386-8567, Japan
| | - Kyoko Matsuo
- Department of Applied Biology, Faculty of Textile Science & Technology, Shinshu University, Ueda, Nagano 386-8567, Japan
| | - Anna F. Wang
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544,
United States
| | - Jesse M. Platt
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544,
United States
| | - Luke H. Bradley
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544,
United States
- Departments of Anatomy & Neurobiology, Molecular & Cellular Biochemistry, Center of Structural Biology, University of Kentucky College of Medicine, Lexington, Kentucky 40536, United States
| | - Michael H. Hecht
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544,
United States
| |
Collapse
|
31
|
Malito E, Bursulaya B, Chen C, Surdo PL, Picchianti M, Balducci E, Biancucci M, Brock A, Berti F, Bottomley MJ, Nissum M, Costantino P, Rappuoli R, Spraggon G. Structural basis for lack of toxicity of the diphtheria toxin mutant CRM197. Proc Natl Acad Sci U S A 2012; 109:5229-34. [PMID: 22431623 PMCID: PMC3325714 DOI: 10.1073/pnas.1201964109] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
CRM197 is an enzymatically inactive and nontoxic form of diphtheria toxin that contains a single amino acid substitution (G52E). Being naturally nontoxic, CRM197 is an ideal carrier protein for conjugate vaccines against encapsulated bacteria and is currently used to vaccinate children globally against Haemophilus influenzae, pneumococcus, and meningococcus. To understand the molecular basis for lack of toxicity in CRM197, we determined the crystal structures of the full-length nucleotide-free CRM197 and of CRM197 in complex with the NAD hydrolysis product nicotinamide (NCA), both at 2.0-Å resolution. The structures show for the first time that the overall fold of CRM197 and DT are nearly identical and that the striking functional difference between the two proteins can be explained by a flexible active-site loop that covers the NAD binding pocket. We present the molecular basis for the increased flexibility of the active-site loop in CRM197 as unveiled by molecular dynamics simulations. These structural insights, combined with surface plasmon resonance, NAD hydrolysis, and differential scanning fluorimetry data, contribute to a comprehensive characterization of the vaccine carrier protein, CRM197.
Collapse
Affiliation(s)
- Enrico Malito
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121
- Novartis Vaccines and Diagnostics, Via Fiorentina 1, 53100 Siena, Italy
| | - Badry Bursulaya
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121
| | - Connie Chen
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121
- Joint Center for Structural Genomics, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121
| | - Paola Lo Surdo
- Novartis Vaccines and Diagnostics, Via Fiorentina 1, 53100 Siena, Italy
| | - Monica Picchianti
- Novartis Vaccines and Diagnostics, Via Fiorentina 1, 53100 Siena, Italy
- Department of Evolutionary Biology, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Enrico Balducci
- School of Biosciences and Biotechnologies, University of Camerino, via Gentile III da Varano, 62032 Camerino, Italy; and
| | - Marco Biancucci
- Novartis Vaccines and Diagnostics, Via Fiorentina 1, 53100 Siena, Italy
- Department of Chemistry, University of Siena, Via A. De Gasperi 2, 53100 Siena, Italy
| | - Ansgar Brock
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121
| | - Francesco Berti
- Novartis Vaccines and Diagnostics, Via Fiorentina 1, 53100 Siena, Italy
| | | | - Mikkel Nissum
- Novartis Vaccines and Diagnostics, Via Fiorentina 1, 53100 Siena, Italy
| | - Paolo Costantino
- Novartis Vaccines and Diagnostics, Via Fiorentina 1, 53100 Siena, Italy
| | - Rino Rappuoli
- Novartis Vaccines and Diagnostics, Via Fiorentina 1, 53100 Siena, Italy
| | - Glen Spraggon
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121
| |
Collapse
|
32
|
Abstract
Telomere DNA-binding proteins protect the ends of chromosomes in eukaryotes. A subset of these proteins are constructed with one or more OB folds and bind with G+T-rich single-stranded DNA found at the extreme termini. The resulting DNA-OB protein complex interacts with other telomere components to coordinate critical telomere functions of DNA protection and DNA synthesis. While the first crystal and NMR structures readily explained protection of telomere ends, the picture of how single-stranded DNA becomes available to serve as primer and template for synthesis of new telomere DNA is only recently coming into focus. New structures of telomere OB fold proteins alongside insights from genetic and biochemical experiments have made significant contributions towards understanding how protein-binding OB proteins collaborate with DNA-binding OB proteins to recruit telomerase and DNA polymerase for telomere homeostasis. This review surveys telomere OB protein structures alongside highly comparable structures derived from replication protein A (RPA) components, with the goal of providing a molecular context for understanding telomere OB protein evolution and mechanism of action in protection and synthesis of telomere DNA.
Collapse
Affiliation(s)
- Martin P Horvath
- Department of Biology, University of Utah, Salt Lake City, Utah 84112-0840, USA.
| |
Collapse
|
33
|
Merlino A, Picone D, Ercole C, Balsamo A, Sica F. Chain termini cross-talk in the swapping process of bovine pancreatic ribonuclease. Biochimie 2012; 94:1108-18. [PMID: 22273774 DOI: 10.1016/j.biochi.2012.01.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 01/11/2012] [Indexed: 10/14/2022]
Abstract
3D domain swapping is the process by which two or more protein molecules exchange part of their structure to form intertwined dimers or higher oligomers. Bovine pancreatic ribonuclease (RNase A) is able to swap the N-terminal α-helix (residues 1-13) and/or the C-terminal β-strand (residues 116-124), thus forming a variety of oligomers, including two different dimers. Cis-trans isomerization of the Asn113-Pro114 peptide group was observed when the protein formed the C-terminal swapped dimer. To study the effect of the substitution of Pro114 on the swapping process of RNase A, we have prepared and characterized the P114A monomeric and dimeric variants of the enzyme. In contrast with previous reports, the crystal structure and NMR data on the monomer reveals a mixed cis-trans conformation for the Asn113-Ala114 peptide group, whereas the X-ray structure of the C-terminal swapped dimer of the variant is very close to that of the corresponding dimer of RNase A. The mutation at the C-terminus affects the capability of the N-terminal α-helix to swap and the stability of both dimeric forms. The present results underscore the importance of the hydration shell in determining the cross-talk between the chain termini in the swapping process of RNase A.
Collapse
Affiliation(s)
- Antonello Merlino
- Department of Chemistry, University of Naples Federico II, Complesso Universitario Monte S. Angelo, Via Cinthia, I-80126 Naples, Italy
| | | | | | | | | |
Collapse
|
34
|
Abstract
Among thousands of homo-oligomeric protein structures, there is a small but growing subset of ‘domain-swapped’ proteins. The term ‘domain swapping,’ originally coined by D. Eisenberg, describes a scenario in which two or more polypeptide chains exchange identical units for oligomerization. This type of assembly could play a role in disease-related aggregation and amyloid formation or as a specific mechanism for regulating function. This chapter introduces terms and features concerning domain swapping, summarizes ideas about its putative mechanisms, reports on domain-swapped structures collected from the literature, and describes a few notable examples in detail.
Collapse
|
35
|
Fontana A, de Laureto PP, Spolaore B, Frare E. Identifying disordered regions in proteins by limited proteolysis. Methods Mol Biol 2012; 896:297-318. [PMID: 22821533 DOI: 10.1007/978-1-4614-3704-8_20] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Limited proteolysis experiments can be successfully used to detect sites of disorder in otherwise folded globular proteins. The approach relies on the fact that the proteolysis of a polypeptide substrate requires its binding in an extended conformation at the protease's active site and thus an enhanced backbone flexibility or local unfolding of the site of proteolytic attack. A striking correlation was found between sites of limited proteolysis and sites of enhanced chain flexibility of the polypeptide chain, this last evaluated by the crystallographically determined B-factor. In numerous cases, it has been shown that limited proteolysis occurs at chain regions characterized by missing electron density and thus being disordered. Therefore, limited proteolysis is a simple and reliable experimental technique that can detect sites of disorder in proteins, thus complementing the results that can be obtained by the use of other physicochemical and computational approaches.
Collapse
Affiliation(s)
- Angelo Fontana
- CRIBI Biotechnology Centre, University of Padua, Padua, Italy.
| | | | | | | |
Collapse
|
36
|
Bray JK, Weiss DR, Levitt M. Optimized torsion-angle normal modes reproduce conformational changes more accurately than cartesian modes. Biophys J 2011; 101:2966-9. [PMID: 22208195 DOI: 10.1016/j.bpj.2011.10.054] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 09/28/2011] [Accepted: 10/31/2011] [Indexed: 11/17/2022] Open
Abstract
We present what to our knowledge is a new method of optimized torsion-angle normal-mode analysis, in which the normal modes move along curved paths in Cartesian space. We show that optimized torsion-angle normal modes reproduce protein conformational changes more accurately than Cartesian normal modes. We also show that orthogonalizing the displacement vectors from torsion-angle normal-mode analysis and projecting them as straight lines in Cartesian space does not lead to better performance than Cartesian normal modes. Clearly, protein motion is more naturally described by curved paths in Cartesian space.
Collapse
Affiliation(s)
- Jenelle K Bray
- Department of Structural Biology, Stanford Medical School, Stanford, California, USA.
| | | | | |
Collapse
|
37
|
Man P, Montagner C, Vitrac H, Kavan D, Pichard S, Gillet D, Forest E, Forge V. Accessibility Changes within Diphtheria Toxin T Domain upon Membrane Penetration Probed by Hydrogen Exchange and Mass Spectrometry. J Mol Biol 2011; 414:123-34. [DOI: 10.1016/j.jmb.2011.09.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 09/26/2011] [Indexed: 01/25/2023]
|
38
|
Shameer K, Shingate PN, Manjunath SCP, Karthika M, Pugalenthi G, Sowdhamini R. 3DSwap: curated knowledgebase of proteins involved in 3D domain swapping. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2011; 2011:bar042. [PMID: 21959866 PMCID: PMC3294423 DOI: 10.1093/database/bar042] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Three-dimensional domain swapping is a unique protein structural phenomenon where two or more protein chains in a protein oligomer share a common structural segment between individual chains. This phenomenon is observed in an array of protein structures in oligomeric conformation. Protein structures in swapped conformations perform diverse functional roles and are also associated with deposition diseases in humans. We have performed in-depth literature curation and structural bioinformatics analyses to develop an integrated knowledgebase of proteins involved in 3D domain swapping. The hallmark of 3D domain swapping is the presence of distinct structural segments such as the hinge and swapped regions. We have curated the literature to delineate the boundaries of these regions. In addition, we have defined several new concepts like ‘secondary major interface’ to represent the interface properties arising as a result of 3D domain swapping, and a new quantitative measure for the ‘extent of swapping’ in structures. The catalog of proteins reported in 3DSwap knowledgebase has been generated using an integrated structural bioinformatics workflow of database searches, literature curation, by structure visualization and sequence–structure–function analyses. The current version of the 3DSwap knowledgebase reports 293 protein structures, the analysis of such a compendium of protein structures will further the understanding molecular factors driving 3D domain swapping. Database URL:http://caps.ncbs.res.in/3dswap
Collapse
Affiliation(s)
- Khader Shameer
- National Centre for Biological Sciences, GKVK Campus, Bangalore, Karnataka, India
| | | | | | | | | | | |
Collapse
|
39
|
On diphtheria toxin fragment A release into the cytosol—Cytochalasin D effect and involvement of actin filaments and eukaryotic elongation factor 2. Int J Biochem Cell Biol 2011; 43:1365-72. [DOI: 10.1016/j.biocel.2011.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 05/13/2011] [Accepted: 05/26/2011] [Indexed: 11/17/2022]
|
40
|
Fratelli F, Abrahão-Neto J, Caricati ATP, Borges MM, Guidolin R, Caricati CP. An alternative method for purifying and detoxifying diphtheria toxin. Toxicon 2011; 57:1093-100. [PMID: 21549738 DOI: 10.1016/j.toxicon.2011.04.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 04/12/2011] [Accepted: 04/19/2011] [Indexed: 11/19/2022]
Abstract
Infections caused by Corynebacterium diphtheriae frequently induce situations in which very small doses of antigens injected intradermally can cause strong inflammatory reactions. This bacterium secretes the diphtheria toxin (DT), a virulence factor that can be lethal to the human organism at doses below 0.1 μg/kg of body weight. The present work proposes alternative methods of DT purification using affinity chromatography and of DT detoxification through conjugating with the polymer methoxypolyethylene glycol activated (mPEG). Tests were performed to evaluate: the formation of edemas and the presence of dermonecrotic activity, in vitro cytotoxicity to Vero cells, the neutralizing activity of serum from guinea pigs immunized with the diphtheria toxoid inactivated with mPEG, and the immunogenic activity of the purified and modified toxin. The results indicated that purification with Blue Sepharose was an efficient method, yielding antigen purity equivalent to 2600 Lf/mg of protein nitrogen. The modification of the Purified Toxin with mPEG did not result in the formation of edema or necrosis although it was immunogenic and stimulated the formation of antibodies that could neutralize the Purified Toxin. The toxoid obtained from the purified toxin maintained its immunogenic characteristics, inducing antibodies with neutralizing activity; edema and necrosis were still observed, however.
Collapse
Affiliation(s)
- Fernando Fratelli
- Lab. of Research and Development of Immunobiologicals for Veterinary Use, Institute Butantan, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
41
|
Mechanism of diphtheria toxin catalytic domain delivery to the eukaryotic cell cytosol and the cellular factors that directly participate in the process. Toxins (Basel) 2011; 3:294-308. [PMID: 22069710 PMCID: PMC3202816 DOI: 10.3390/toxins3030294] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 03/03/2011] [Accepted: 03/10/2011] [Indexed: 11/17/2022] Open
Abstract
Research on diphtheria and anthrax toxins over the past three decades has culminated in a detailed understanding of their structure function relationships (e.g., catalytic (C), transmembrane (T), and receptor binding (R) domains), as well as the identification of their eukaryotic cell surface receptor, an understanding of the molecular events leading to the receptor-mediated internalization of the toxin into an endosomal compartment, and the pH triggered conformational changes required for pore formation in the vesicle membrane. Recently, a major research effort has been focused on the development of a detailed understanding of the molecular interactions between each of these toxins and eukaryotic cell factors that play an essential role in the efficient translocation of their respective catalytic domains through the trans-endosomal vesicle membrane pore and delivery into the cell cytosol. In this review, I shall focus on recent findings that have led to a more detailed understanding of the mechanism by which the diphtheria toxin catalytic domain is delivered to the eukaryotic cell cytosol. While much work remains, it is becoming increasingly clear that the entry process is facilitated by specific interactions with a number of cellular factors in an ordered sequential fashion. In addition, since diphtheria, anthrax lethal factor and anthrax edema factor all carry multiple coatomer I complex binding motifs and COPI complex has been shown to play an essential role in entry process, it is likely that the initial steps in catalytic domain entry of these divergent toxins follow a common mechanism.
Collapse
|
42
|
Chassaing A, Pichard S, Araye-Guet A, Barbier J, Forge V, Gillet D. Solution and membrane-bound chaperone activity of the diphtheria toxin translocation domain towards the catalytic domain. FEBS J 2011; 278:4516-25. [PMID: 21332941 DOI: 10.1111/j.1742-4658.2011.08053.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
During cell intoxication by diphtheria toxin, endosome acidification triggers the translocation of the catalytic (C) domain into the cytoplasm. This event is mediated by the translocation (T) domain of the toxin. Previous work suggested that the T domain acts as a chaperone for the C domain during membrane penetration of the toxin. Using partitioning experiments with lipid vesicles, fluorescence spectroscopy, and a lipid vesicle leakage assay, we characterized the dominant behavior of the T domain over the C domain during the successive steps by which these domains interact with a membrane upon acidification: partial unfolding in solution and during membrane binding, and then structural rearrangement during penetration into the membrane. To this end, we compared, for each domain, isolated or linked together in a CT protein (the toxin lacking the receptor-binding domain), each of these steps. The behavior of the T domain is marginally modified by the presence or absence of the C domain, whereas that of the C domain is greatly affected by the presence of the T domain . All of the steps leading to membrane penetration of the C domain are triggered at higher pH by the T domain , by 0.5-1.6 pH units. The T domain stabilizes the partially folded states of the C domain corresponding to each step of the process. The results unambiguously demonstrate that the T domain acts as a specialized pH-dependent chaperone for the C domain. Interestingly, this chaperone activity acts on very different states of the protein: in solution, membrane-bound, and membrane-inserted.
Collapse
Affiliation(s)
- Anne Chassaing
- Commissariat à l'Energie Atomique (CEA), Institut de Biologie et Technologies de Saclay (iBiTecS), Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), Gif sur Yvette, France
| | | | | | | | | | | |
Collapse
|
43
|
Chu CH, Lo WC, Wang HW, Hsu YC, Hwang JK, Lyu PC, Pai TW, Tang CY. Detection and alignment of 3D domain swapping proteins using angle-distance image-based secondary structural matching techniques. PLoS One 2010; 5:e13361. [PMID: 20976204 PMCID: PMC2955075 DOI: 10.1371/journal.pone.0013361] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2010] [Accepted: 09/13/2010] [Indexed: 11/18/2022] Open
Abstract
This work presents a novel detection method for three-dimensional domain swapping (DS), a mechanism for forming protein quaternary structures that can be visualized as if monomers had “opened” their “closed” structures and exchanged the opened portion to form intertwined oligomers. Since the first report of DS in the mid 1990s, an increasing number of identified cases has led to the postulation that DS might occur in a protein with an unconstrained terminus under appropriate conditions. DS may play important roles in the molecular evolution and functional regulation of proteins and the formation of depositions in Alzheimer's and prion diseases. Moreover, it is promising for designing auto-assembling biomaterials. Despite the increasing interest in DS, related bioinformatics methods are rarely available. Owing to a dramatic conformational difference between the monomeric/closed and oligomeric/open forms, conventional structural comparison methods are inadequate for detecting DS. Hence, there is also a lack of comprehensive datasets for studying DS. Based on angle-distance (A-D) image transformations of secondary structural elements (SSEs), specific patterns within A-D images can be recognized and classified for structural similarities. In this work, a matching algorithm to extract corresponding SSE pairs from A-D images and a novel DS score have been designed and demonstrated to be applicable to the detection of DS relationships. The Matthews correlation coefficient (MCC) and sensitivity of the proposed DS-detecting method were higher than 0.81 even when the sequence identities of the proteins examined were lower than 10%. On average, the alignment percentage and root-mean-square distance (RMSD) computed by the proposed method were 90% and 1.8Å for a set of 1,211 DS-related pairs of proteins. The performances of structural alignments remain high and stable for DS-related homologs with less than 10% sequence identities. In addition, the quality of its hinge loop determination is comparable to that of manual inspection. This method has been implemented as a web-based tool, which requires two protein structures as the input and then the type and/or existence of DS relationships between the input structures are determined according to the A-D image-based structural alignments and the DS score. The proposed method is expected to trigger large-scale studies of this interesting structural phenomenon and facilitate related applications.
Collapse
Affiliation(s)
- Chia-Han Chu
- Department of Computer Science, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Wei-Cheng Lo
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan, Republic of China
| | - Hsin-Wei Wang
- Department of Computer Science and Engineering, National Taiwan Ocean University, Keelung, Taiwan, Republic of China
| | - Yen-Chu Hsu
- Department of Computer Science and Engineering, National Taiwan Ocean University, Keelung, Taiwan, Republic of China
| | - Jenn-Kang Hwang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan, Republic of China
| | - Ping-Chiang Lyu
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Tun-Wen Pai
- Department of Computer Science and Engineering, National Taiwan Ocean University, Keelung, Taiwan, Republic of China
- * E-mail: (T-WP); (CYT)
| | - Chuan Yi Tang
- Department of Computer Science, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
- Department of Computer Science and Information Engineering, Providence University, Taichung, Taiwan, Republic of China
- * E-mail: (T-WP); (CYT)
| |
Collapse
|
44
|
Abstract
Thioredoxins are ubiquitous antioxidant enzymes that play important roles in many health-related cellular processes. As such, the fundamental knowledge of how these enzymes work is of prime importance for understanding cellular redox mechanisms and for laying the ground for the development of future therapeutic approaches. Over the past 40 years, a really impressive amount of data has been published on thioredoxins. Here, we review the most significant results that have contributed to our knowledge regarding the structure, the function, and the mechanism of these crucial enzymes.
Collapse
|
45
|
Shameer K, Pugalenthi G, Kandaswamy KK, Suganthan PN, Archunan G, Sowdhamini R. Insights into Protein Sequence and Structure-Derived Features Mediating 3D Domain Swapping Mechanism using Support Vector Machine Based Approach. Bioinform Biol Insights 2010; 4:33-42. [PMID: 20634983 PMCID: PMC2901629 DOI: 10.4137/bbi.s4464] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
3-dimensional domain swapping is a mechanism where two or more protein molecules form higher order oligomers by exchanging identical or similar subunits. Recently, this phenomenon has received much attention in the context of prions and neurodegenerative diseases, due to its role in the functional regulation, formation of higher oligomers, protein misfolding, aggregation etc. While 3-dimensional domain swap mechanism can be detected from three-dimensional structures, it remains a formidable challenge to derive common sequence or structural patterns from proteins involved in swapping. We have developed a SVM-based classifier to predict domain swapping events using a set of features derived from sequence and structural data. The SVM classifier was trained on features derived from 150 proteins reported to be involved in 3D domain swapping and 150 proteins not known to be involved in swapped conformation or related to proteins involved in swapping phenomenon. The testing was performed using 63 proteins from the positive dataset and 63 proteins from the negative dataset. We obtained 76.33% accuracy from training and 73.81% accuracy from testing. Due to high diversity in the sequence, structure and functions of proteins involved in domain swapping, availability of such an algorithm to predict swapping events from sequence and structure-derived features will be an initial step towards identification of more putative proteins that may be involved in swapping or proteins involved in deposition disease. Further, the top features emerging in our feature selection method may be analysed further to understand their roles in the mechanism of domain swapping.
Collapse
Affiliation(s)
- Khader Shameer
- National Centre for Biological Sciences (TIFR), GKVK Campus, Bellary Road, Bangalore, 560065, India
| | | | | | | | | | | |
Collapse
|
46
|
Nagarkar RP, Hule RA, Pochan DJ, Schneider JP. Domain swapping in materials design. Biopolymers 2010; 94:141-55. [PMID: 20091872 DOI: 10.1002/bip.21332] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Peptide self-assembly can be used as a bottom-up approach to material fabrication. Although many different types of materials can be prepared from peptides, hydrogels are perhaps one of the most common. Gels typically result from the self-assembly of peptides into fibrillar networks. Controlling the structural morphology of these fibrils and the networks they form allows direct control over a given material's bulk properties. However, exerting this control is extremely difficult as the mechanistic rules that govern peptide self-assembly are far from being established. Conversely, several amyloidogenic proteins have been shown to self-assemble into fibrils using a mechanism known as domain swapping. Here, discrete units of secondary structure or even whole domains are exchanged (swapped) among discrete proteins during self-assembly to form extended networks with precise structural control. This review discusses several common mechanistic variations of domain swapping using naturally occurring proteins as examples. The possibility of using these principles to design peptides capable of controlled assembly and fibril formation leading to materials with targeted properties is explored.
Collapse
Affiliation(s)
- Radhika P Nagarkar
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | | | | | | |
Collapse
|
47
|
Esposito V, Guglielmi F, Martin SR, Pauwels K, Pastore A, Piccoli R, Temussi PA. Aggregation Mechanisms of Cystatins: A Comparative Study of Monellin and Oryzacystatin. Biochemistry 2010; 49:2805-10. [DOI: 10.1021/bi902039s] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Veronica Esposito
- MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, U.K
| | - Fulvio Guglielmi
- Department of Structural and Functional Biology, University of Naples “Federico II”, School of Biotechnology, Naples, Italy
| | - Stephen R. Martin
- MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, U.K
| | - Kris Pauwels
- MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, U.K
| | - Annalisa Pastore
- MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, U.K
| | - Renata Piccoli
- Department of Structural and Functional Biology, University of Naples “Federico II”, School of Biotechnology, Naples, Italy
- Istituto Nazionale di Biostrutture e Biosistemi (INBB), Naples, Italy
| | - Piero A. Temussi
- MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, U.K
- Department of Chemistry, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
48
|
Wouters MA, Fan SW, Haworth NL. Disulfides as redox switches: from molecular mechanisms to functional significance. Antioxid Redox Signal 2010; 12:53-91. [PMID: 19634988 DOI: 10.1089/ars.2009.2510] [Citation(s) in RCA: 182] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The molecular mechanisms underlying thiol-based redox control are poorly defined. Disulfide bonds between Cys residues are commonly thought to confer extra rigidity and stability to their resident protein, forming a type of proteinaceous spot weld. Redox biologists have been redefining the role of disulfides over the last 30-40 years. Disulfides are now known to form in the cytosol under conditions of oxidative stress. Isomerization of extracellular disulfides is also emerging as an important regulator of protein function. The current paradigm is that the disulfide proteome consists of two subproteomes: a structural group and a redox-sensitive group. The redox-sensitive group is less stable and often associated with regions of stress in protein structures. Some characterized redox-active disulfides are the helical CXXC motif, often associated with thioredoxin-fold proteins; and forbidden disulfides, a group of metastable disulfides that disobey elucidated rules of protein stereochemistry. Here we discuss the role of redox-active disulfides as switches in proteins.
Collapse
Affiliation(s)
- Merridee A Wouters
- Structural & Computational Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia.
| | | | | |
Collapse
|
49
|
Man P, Montagner C, Vitrac H, Kavan D, Pichard S, Gillet D, Forest E, Forge V. Accessibility changes within diphtheria toxin T domain when in the functional molten globule state, as determined using hydrogen/deuterium exchange measurements. FEBS J 2009; 277:653-62. [PMID: 20050921 DOI: 10.1111/j.1742-4658.2009.07511.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The translocation domain (T domain) of diphtheria toxin adopts a partially folded state, the so-called molten globule state, to become functional at acidic pH. We compared, using hydrogen/deuterium exchange experiments associated with MS, the structures of the T domain in its soluble folded state at neutral pH and in its functional molten globule state at acidic pH. In the native state, the alpha-helices TH5 and TH8 are identified as the core of the domain. Based on the high-resolution structure of the T domain, we propose that TH8 is highly protected because it is buried within the native structure. According to the same structure, TH5 is partly accessible at the surface of the T domain. We propose that its high protection is caused by the formation of dimers. Within the molten globule state, high protection is still observed within the helical hairpin TH8-TH9, which is responsible for the insertion of the T domain into the membrane. In the absence of the lipid bilayer, this hydrophobic part of the domain self-assembles, leading to the formation of oligomers. Overall, hydrogen/deuterium-exchange measurements allow the analysis of interaction contacts within small oligomers made of partially folded proteins. Such information, together with crystal structure data, are particularly valuable for using to analyze the self-assembly of proteins.
Collapse
Affiliation(s)
- Petr Man
- Laboratoire de Spectrométrie de Masse des Protéines, Institut de Biologie Structurale (CEA, CNRS, UJF, UMR 5075), Grenoble, France
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Ercole C, Colamarino RA, Pizzo E, Fogolari F, Spadaccini R, Picone D. Comparison of the structural and functional properties of RNase A and BS-RNase: A stepwise mutagenesis approach. Biopolymers 2009; 91:1009-17. [DOI: 10.1002/bip.21176] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|