1
|
Petersen M, Behera SP, Majumdar A, Barrick D. Thermodynamic Coupling in a Consensus-Designed Spectrin Repeat Protein. J Phys Chem B 2025; 129:4614-4628. [PMID: 40324019 DOI: 10.1021/acs.jpcb.4c08772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Cooperativity is a central feature to protein folding and is important for protein design. Repeat proteins are good systems for quantifying the thermodynamic basis of cooperativity. Analysis of repeat proteins composed of identical consensus repeats show that repeats strongly drive the folding of their neighbors through extensive tertiary contacts. Here, we use the consensus approach to quantify the cooperativity of folding of spectrin repeat arrays. Spectrin repeats are unique among tandem repeat proteins in that they share an elongated α-helix that spans neighboring repeats. We generate a consensus spectrin repeat sequence and show that this sequence is structured by CD and NMR spectroscopy, and is considerably more stable than extant spectrin repeats. By generating pairs of consensus spectrin repeats, we find tandem repeats to be further stabilized, demonstrating cooperative stabilization by neighboring repeats. Using an Ising model to analyze single- and tandem spectrin repeat unfolding, we find that the consensus stability increase results from intrinsic but not interfacial stabilization. By introducing mutations and insertions at the boundary between consensus repeats, we find that cooperativity is driven primarily by helical propagation; to a lesser extent, helix propagation also stabilizes partly folded states where one of two repeats is unfolded.
Collapse
Affiliation(s)
- Mark Petersen
- The T.C. Jenkins Department of Biophysics, Johns Hopkins University, 3400 N. Charles St., Baltimore, Maryland 21218, United States
| | - Soumya Prakash Behera
- The T.C. Jenkins Department of Biophysics, Johns Hopkins University, 3400 N. Charles St., Baltimore, Maryland 21218, United States
| | - Ananya Majumdar
- The Biophysical NMR Center, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Doug Barrick
- The T.C. Jenkins Department of Biophysics, Johns Hopkins University, 3400 N. Charles St., Baltimore, Maryland 21218, United States
| |
Collapse
|
2
|
Du M, Zeng F, Wang Y, Li Y, Chen G, Jiang J, Wang Q. Assembly and Functionality of 2D Protein Arrays. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416485. [PMID: 40089855 PMCID: PMC12005781 DOI: 10.1002/advs.202416485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/16/2025] [Indexed: 03/17/2025]
Abstract
Among the unique classes of 2D nanomaterials, 2D protein arrays garner increasing attention due to their remarkable structural stability, exceptional physiochemical properties, and tunable electronic and mechanical attributes. The interest in mimicking and surpassing the precise architecture and advanced functionality of natural protein systems drives the field of 2D protein assembly toward the development of sophisticated functional materials. Recent advancements deepen the understanding of the fundamental principles governing 2D protein self-assembly, accelerating the creation of novel functional biomaterials. These developments encompass biological, chemical, and templated strategies, facilitating the self-organization of proteins into highly ordered and intricate 2D patterns. Consequently, these 2D protein arrays create new opportunities for integrating diverse components, from small molecules to nanoparticles, thereby enhancing the performance and versatility of materials in various applications. This review comprehensively assesses the current state of 2D protein nanotechnology, highlighting the latest methodologies for directing protein assembly into precise 2D architectures. The transformative potential of 2D protein assemblies in designing next-generation biomaterials, particularly in areas such as biomedicine, catalysis, photosystems, and membrane filtration is also emphasized.
Collapse
Affiliation(s)
- Mingming Du
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Fanmeng Zeng
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - YueFei Wang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Ying Li
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Guangcun Chen
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Jiang Jiang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Qiangbin Wang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Physical Science and TechnologyShanghaiTech UniversityShanghai201210China
- College of Materials Sciences and Opto‐Electronic TechnologyUniversity of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
3
|
Zhang T, Yang DB, Kloxin CJ, Pochan DJ, Saven JG. Coarse-Grain Model of Ultrarigid Polymer Rods Comprising Bifunctionally Linked Peptide Bundlemers. Biomacromolecules 2024; 25:7904-7914. [PMID: 39499090 DOI: 10.1021/acs.biomac.4c01192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Computationally designed homotetrameric helical peptide bundles have been functionalized at their N-termini to achieve supramolecular polymers, wherein individual bundles ("bundlemers") are the monomeric units. Adjacent bundles are linked via two covalent cross-links. The polymers exhibit a range of conformational properties, including formation of rigid-rods with micrometer-scale persistence lengths. Herein, a coarse-grained model is used to illuminate how molecular features affect the rod-like behavior of the polymers. With increasing affinity between bundlemer ends, a sharp transition in the persistence length is observed. Doubly linked chains exhibit larger persistence lengths and more robust formation of rigid-rod structures than singly linked chains. Chain stiffness increases with decreasing temperatures. Increasing the length of the cross-linker results in more flexible chains. This model provides insights into how molecular features control the structural properties of chains comprising doubly linked rigid bundlemers.
Collapse
Affiliation(s)
- Tianren Zhang
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Dai-Bei Yang
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Christopher J Kloxin
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Darrin J Pochan
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Jeffery G Saven
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
4
|
Melnik TN, Majorina MA, Vorobeva DE, Nagibina GS, Veselova VR, Glukhova KA, Pak MA, Ivankov DN, Uversky VN, Melnik BS. Design of stable circular permutants of the GroEL chaperone apical domain. Cell Commun Signal 2024; 22:90. [PMID: 38303060 PMCID: PMC10836027 DOI: 10.1186/s12964-023-01426-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/08/2023] [Indexed: 02/03/2024] Open
Abstract
Enhancing protein stability holds paramount significance in biotechnology, therapeutics, and the food industry. Circular permutations offer a distinctive avenue for manipulating protein stability while keeping intra-protein interactions intact. Amidst the creation of circular permutants, determining the optimal placement of the new N- and C-termini stands as a pivotal, albeit largely unexplored, endeavor. In this study, we employed PONDR-FIT's predictions of disorder propensity to guide the design of circular permutants for the GroEL apical domain (residues 191-345). Our underlying hypothesis posited that a higher predicted disorder value would correspond to reduced stability in the circular permutants, owing to the increased likelihood of fluctuations in the novel N- and C-termini. To substantiate this hypothesis, we engineered six circular permutants, positioning glycines within the loops as locations for the new N- and C-termini. We demonstrated the validity of our hypothesis along the set of the designed circular permutants, as supported by measurements of melting temperatures by circular dichroism and differential scanning microcalorimetry. Consequently, we propose a novel computational methodology that rationalizes the design of circular permutants with projected stability. Video Abstract.
Collapse
Affiliation(s)
- Tatiana N Melnik
- Institute of Protein Research, Russian Academy of Sciences, Institutskaja Str. 4, Pushchino, Moscow Region, 142290, Russia
| | - Maria A Majorina
- Institute of Protein Research, Russian Academy of Sciences, Institutskaja Str. 4, Pushchino, Moscow Region, 142290, Russia
| | - Daria E Vorobeva
- Institute of Protein Research, Russian Academy of Sciences, Institutskaja Str. 4, Pushchino, Moscow Region, 142290, Russia
| | - Galina S Nagibina
- Institute of Protein Research, Russian Academy of Sciences, Institutskaja Str. 4, Pushchino, Moscow Region, 142290, Russia
| | - Victoria R Veselova
- Institute of Protein Research, Russian Academy of Sciences, Institutskaja Str. 4, Pushchino, Moscow Region, 142290, Russia
| | - Ksenia A Glukhova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaja Str. 3, Puschino, Moscow Region, 142290, Russia
| | - Marina A Pak
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Bld. 1, Moscow, 121205, Russia
| | - Dmitry N Ivankov
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Bld. 1, Moscow, 121205, Russia
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Center and Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia.
| | - Bogdan S Melnik
- Institute of Protein Research, Russian Academy of Sciences, Institutskaja Str. 4, Pushchino, Moscow Region, 142290, Russia.
- Pushchino Branch, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki 6, Pushchino, Moscow Region, 142290, Russia.
| |
Collapse
|
5
|
Xu B, Chen Y, Xue W. Computational Protein Design - Where it goes? Curr Med Chem 2024; 31:2841-2854. [PMID: 37272467 DOI: 10.2174/0929867330666230602143700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 02/18/2023] [Accepted: 03/15/2023] [Indexed: 06/06/2023]
Abstract
Proteins have been playing a critical role in the regulation of diverse biological processes related to human life. With the increasing demand, functional proteins are sparse in this immense sequence space. Therefore, protein design has become an important task in various fields, including medicine, food, energy, materials, etc. Directed evolution has recently led to significant achievements. Molecular modification of proteins through directed evolution technology has significantly advanced the fields of enzyme engineering, metabolic engineering, medicine, and beyond. However, it is impossible to identify desirable sequences from a large number of synthetic sequences alone. As a result, computational methods, including data-driven machine learning and physics-based molecular modeling, have been introduced to protein engineering to produce more functional proteins. This review focuses on recent advances in computational protein design, highlighting the applicability of different approaches as well as their limitations.
Collapse
Affiliation(s)
- Binbin Xu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yingjun Chen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Weiwei Xue
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| |
Collapse
|
6
|
Padhi AK, Kalita P, Maurya S, Poluri KM, Tripathi T. From De Novo Design to Redesign: Harnessing Computational Protein Design for Understanding SARS-CoV-2 Molecular Mechanisms and Developing Therapeutics. J Phys Chem B 2023; 127:8717-8735. [PMID: 37815479 DOI: 10.1021/acs.jpcb.3c04542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
The continuous emergence of novel SARS-CoV-2 variants and subvariants serves as compelling evidence that COVID-19 is an ongoing concern. The swift, well-coordinated response to the pandemic highlights how technological advancements can accelerate the detection, monitoring, and treatment of the disease. Robust surveillance systems have been established to understand the clinical characteristics of new variants, although the unpredictable nature of these variants presents significant challenges. Some variants have shown resistance to current treatments, but innovative technologies like computational protein design (CPD) offer promising solutions and versatile therapeutics against SARS-CoV-2. Advances in computing power, coupled with open-source platforms like AlphaFold and RFdiffusion (employing deep neural network and diffusion generative models), among many others, have accelerated the design of protein therapeutics with precise structures and intended functions. CPD has played a pivotal role in developing peptide inhibitors, mini proteins, protein mimics, decoy receptors, nanobodies, monoclonal antibodies, identifying drug-resistance mutations, and even redesigning native SARS-CoV-2 proteins. Pending regulatory approval, these designed therapies hold the potential for a lasting impact on human health and sustainability. As SARS-CoV-2 continues to evolve, use of such technologies enables the ongoing development of alternative strategies, thus equipping us for the "New Normal".
Collapse
Affiliation(s)
- Aditya K Padhi
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Parismita Kalita
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North-Eastern Hill University, Shillong 793022, India
| | - Shweata Maurya
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Timir Tripathi
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North-Eastern Hill University, Shillong 793022, India
- Department of Zoology, School of Life Sciences, North-Eastern Hill University, Shillong 793022, India
| |
Collapse
|
7
|
Ansbacher T, Tohar R, Cohen A, Cohen O, Levartovsky S, Arieli A, Matalon S, Bar DZ, Gal M, Weinberg E. A novel computationally engineered collagenase reduces the force required for tooth extraction in an ex-situ porcine jaw model. J Biol Eng 2023; 17:47. [PMID: 37461028 DOI: 10.1186/s13036-023-00366-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 07/05/2023] [Indexed: 07/20/2023] Open
Abstract
The currently employed tooth extraction methods in dentistry involve mechanical disruption of the periodontal ligament fibers, leading to inevitable trauma to the bundle bone comprising the socket walls. In our previous work, we have shown that a recombinantly expressed truncated version of clostridial collagenase G (ColG) purified from Escherichia coli efficiently reduced the force needed for tooth extraction in an ex-situ porcine jaw model, when injected into the periodontal ligament. Considering that enhanced thermostability often leads to higher enzymatic activity and to set the basis for additional rounds of optimization, we used a computational protein design approach to generate an enzyme to be more thermostable while conserving the key catalytic residues. This process generated a novel collagenase (ColG-variant) harboring sixteen mutations compared to ColG, with a nearly 4℃ increase in melting temperature. Herein, we explored the potential of ColG-variant to further decrease the physical effort required for tooth delivery using our established ex-situ porcine jaw model. An average reduction of 11% was recorded in the force applied to extract roots of mandibular split first and second premolar teeth treated with ColG-variant, relative to those treated with ColG. Our results show for the first time the potential of engineering enzyme properties for dental medicine and further contribute to minimally invasive tooth extraction.
Collapse
Affiliation(s)
- Tamar Ansbacher
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
- Hadassah Academic College, 91010, Jerusalem, Israel
| | - Ran Tohar
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Adi Cohen
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Orel Cohen
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Shifra Levartovsky
- Department of Oral Rehabilitation, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Adi Arieli
- Department of Oral Rehabilitation, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Shlomo Matalon
- Department of Oral Rehabilitation, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Daniel Z Bar
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Maayan Gal
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.
| | - Evgeny Weinberg
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.
- Department of Periodontology and Oral Implantology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.
| |
Collapse
|
8
|
Thakkar R, Upreti D, Ishiguro S, Tamura M, Comer J. Computational design of a cyclic peptide that inhibits the CTLA4 immune checkpoint. RSC Med Chem 2023; 14:658-670. [PMID: 37122540 PMCID: PMC10131585 DOI: 10.1039/d2md00409g] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Proteins involved in immune checkpoint pathways, such as CTLA4, PD1, and PD-L1, have become important targets for cancer immunotherapy; however, development of small molecule drugs targeting these pathways has proven difficult due to the nature of their protein-protein interfaces. Here, using a hierarchy of computational techniques, we design a cyclic peptide that binds CTLA4 and follow this with experimental verification of binding and biological activity, using bio-layer interferometry, cell culture, and a mouse tumor model. Beginning from a template excised from the X-ray structure of the CTLA4:B7-2 complex, we generate several peptide sequences using flexible docking and modeling steps. These peptides are cyclized head-to-tail to improve structural and proteolytic stability and screened using molecular dynamics simulation and MM-GBSA calculation. The standard binding free energies for shortlisted peptides are then calculated in explicit-solvent simulation using a rigorous multistep technique. The most promising peptide, cyc(EIDTVLTPTGWVAKRYS), yields the standard free energy -6.6 ± 3.5 kcal mol-1, which corresponds to a dissociation constant of ∼15 μmol L-1. The binding affinity of this peptide for CTLA4 is measured experimentally (31 ± 4 μmol L-1) using bio-layer interferometry. Treatment with this peptide inhibited tumor growth in a co-culture of Lewis lung carcinoma (LLC) cells and antigen primed T cells, as well as in mice with an orthotropic Lewis lung carcinoma allograft model.
Collapse
Affiliation(s)
- Ravindra Thakkar
- Department of Anatomy and Physiology, Kansas State University 1620 Denison Avenue Manhattan Kansas USA +1 785 532 6311
| | - Deepa Upreti
- Department of Anatomy and Physiology, Kansas State University 1620 Denison Avenue Manhattan Kansas USA +1 785 532 6311
| | - Susumu Ishiguro
- Department of Anatomy and Physiology, Kansas State University 1620 Denison Avenue Manhattan Kansas USA +1 785 532 6311
| | - Masaaki Tamura
- Department of Anatomy and Physiology, Kansas State University 1620 Denison Avenue Manhattan Kansas USA +1 785 532 6311
| | - Jeffrey Comer
- Department of Anatomy and Physiology, Kansas State University 1620 Denison Avenue Manhattan Kansas USA +1 785 532 6311
| |
Collapse
|
9
|
Runthala A, Mbye M, Ayyash M, Xu Y, Kamal-Eldin A. Caseins: Versatility of Their Micellar Organization in Relation to the Functional and Nutritional Properties of Milk. Molecules 2023; 28:molecules28052023. [PMID: 36903269 PMCID: PMC10004547 DOI: 10.3390/molecules28052023] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/24/2023] Open
Abstract
The milk of mammals is a complex fluid mixture of various proteins, minerals, lipids, and other micronutrients that play a critical role in providing nutrition and immunity to newborns. Casein proteins together with calcium phosphate form large colloidal particles, called casein micelles. Caseins and their micelles have received great scientific interest, but their versatility and role in the functional and nutritional properties of milk from different animal species are not fully understood. Caseins belong to a class of proteins that exhibit open and flexible conformations. Here, we discuss the key features that maintain the structures of the protein sequences in four selected animal species: cow, camel, human, and African elephant. The primary sequences of these proteins and their posttranslational modifications (phosphorylation and glycosylation) that determine their secondary structures have distinctively evolved in these different animal species, leading to differences in their structural, functional, and nutritional properties. The variability in the structures of milk caseins influence the properties of their dairy products, such as cheese and yogurt, as well as their digestibility and allergic properties. Such differences are beneficial to the development of different functionally improved casein molecules with variable biological and industrial utilities.
Collapse
Affiliation(s)
- Ashish Runthala
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Vijayawada 522302, India
- Correspondence: (A.R.); (A.K.-E.); Tel.: +971-5-0138-9248 (A.K.-E.)
| | - Mustapha Mbye
- Department of Food Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Mutamed Ayyash
- Department of Food Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Yajun Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100871, China
| | - Afaf Kamal-Eldin
- Department of Food Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Correspondence: (A.R.); (A.K.-E.); Tel.: +971-5-0138-9248 (A.K.-E.)
| |
Collapse
|
10
|
Wang X, Xu K, Tan Y, Liu S, Zhou J. Possibilities of Using De Novo Design for Generating Diverse Functional Food Enzymes. Int J Mol Sci 2023; 24:3827. [PMID: 36835238 PMCID: PMC9964944 DOI: 10.3390/ijms24043827] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 02/17/2023] Open
Abstract
Food enzymes have an important role in the improvement of certain food characteristics, such as texture improvement, elimination of toxins and allergens, production of carbohydrates, enhancing flavor/appearance characteristics. Recently, along with the development of artificial meats, food enzymes have been employed to achieve more diverse functions, especially in converting non-edible biomass to delicious foods. Reported food enzyme modifications for specific applications have highlighted the significance of enzyme engineering. However, using direct evolution or rational design showed inherent limitations due to the mutation rates, which made it difficult to satisfy the stability or specific activity needs for certain applications. Generating functional enzymes using de novo design, which highly assembles naturally existing enzymes, provides potential solutions for screening desired enzymes. Here, we describe the functions and applications of food enzymes to introduce the need for food enzymes engineering. To illustrate the possibilities of using de novo design for generating diverse functional proteins, we reviewed protein modelling and de novo design methods and their implementations. The future directions for adding structural data for de novo design model training, acquiring diversified training data, and investigating the relationship between enzyme-substrate binding and activity were highlighted as challenges to overcome for the de novo design of food enzymes.
Collapse
Affiliation(s)
- Xinglong Wang
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Kangjie Xu
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Yameng Tan
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Song Liu
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Jingwen Zhou
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
11
|
Lu H, Cheng Z, Hu Y, Tang LV. What Can De Novo Protein Design Bring to the Treatment of Hematological Disorders? BIOLOGY 2023; 12:166. [PMID: 36829445 PMCID: PMC9952452 DOI: 10.3390/biology12020166] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Protein therapeutics have been widely used to treat hematological disorders. With the advent of de novo protein design, protein therapeutics are not limited to ameliorating natural proteins but also produce novel protein sequences, folds, and functions with shapes and functions customized to bind to the therapeutic targets. De novo protein techniques have been widely used biomedically to design novel diagnostic and therapeutic drugs, novel vaccines, and novel biological materials. In addition, de novo protein design has provided new options for treating hematological disorders. Scientists have designed protein switches called Colocalization-dependent Latching Orthogonal Cage-Key pRoteins (Co-LOCKR) that perform computations on the surface of cells. De novo designed molecules exhibit a better capacity than the currently available tyrosine kinase inhibitors in chronic myeloid leukemia therapy. De novo designed protein neoleukin-2/15 enhances chimeric antigen receptor T-cell activity. This new technique has great biomedical potential, especially in exploring new treatment methods for hematological disorders. This review discusses the development of de novo protein design and its biological applications, with emphasis on the treatment of hematological disorders.
Collapse
Affiliation(s)
| | | | | | - Liang V. Tang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
12
|
Ribeiro AL, Sánchez M, Bosch S, Berenguer J, Hidalgo A. Stabilization of Enzymes by Using Thermophiles. Methods Mol Biol 2023; 2704:313-328. [PMID: 37642853 DOI: 10.1007/978-1-0716-3385-4_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Manufactured steroid compounds have many applications in the pharmaceutical industry. Due to the chemical complexity and chirality of steroids, there is an increasing demand for enzyme-based bioconversion processes to replace those based on chemical synthesis. In this context, thermostability of the involved enzymes is a highly desirable property as both the increased half-life of the enzyme and the enhanced solubility of substrates and products will improve the yield of the reactions. Metagenomic libraries from thermal environments are potential sources of thermostable enzymes of prokaryotic origin, but the number of expected hits could be quite low for enzymes handling substrates such as steroids, rarely found in prokaryotes. An alternative to metagenome screening is the selection of thermostable variants of well-known steroid-processing enzymes. Here we review and detail a protocol for such selection, where error-prone PCR (epPCR) is used to introduce random mutations into a gene to create a variants library for further selection of thermostable variants in the thermophile Thermus thermophilus. The method involves the use of folding interference vectors where the proper folding of the enzyme of interest at high temperature is linked to the folding of a reporter encoding a selectable property such as thermostable resistance to kanamycin, leading to a life-or-death selection of variants of reinforced folding independently of the activity of the enzyme.
Collapse
Affiliation(s)
- Ana-Luisa Ribeiro
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC). Facultad de Ciencias. Universidad Autónoma de Madrid, Madrid, Spain
| | - Mercedes Sánchez
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC). Facultad de Ciencias. Universidad Autónoma de Madrid, Madrid, Spain
| | - Sandra Bosch
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC). Facultad de Ciencias. Universidad Autónoma de Madrid, Madrid, Spain
| | - José Berenguer
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC). Facultad de Ciencias. Universidad Autónoma de Madrid, Madrid, Spain
| | - Aurelio Hidalgo
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC). Facultad de Ciencias. Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
13
|
Dutta P, Roy P, Sengupta N. Effects of External Perturbations on Protein Systems: A Microscopic View. ACS OMEGA 2022; 7:44556-44572. [PMID: 36530249 PMCID: PMC9753117 DOI: 10.1021/acsomega.2c06199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/14/2022] [Indexed: 06/17/2023]
Abstract
Protein folding can be viewed as the origami engineering of biology resulting from the long process of evolution. Even decades after its recognition, research efforts worldwide focus on demystifying molecular factors that underlie protein structure-function relationships; this is particularly relevant in the era of proteopathic disease. A complex co-occurrence of different physicochemical factors such as temperature, pressure, solvent, cosolvent, macromolecular crowding, confinement, and mutations that represent realistic biological environments are known to modulate the folding process and protein stability in unique ways. In the current review, we have contextually summarized the substantial efforts in unveiling individual effects of these perturbative factors, with major attention toward bottom-up approaches. Moreover, we briefly present some of the biotechnological applications of the insights derived from these studies over various applications including pharmaceuticals, biofuels, cryopreservation, and novel materials. Finally, we conclude by summarizing the challenges in studying the combined effects of multifactorial perturbations in protein folding and refer to complementary advances in experiment and computational techniques that lend insights to the emergent challenges.
Collapse
Affiliation(s)
- Pallab Dutta
- Department
of Biological Sciences, Indian Institute
of Science Education and Research (IISER) Kolkata, Mohanpur741246, India
| | - Priti Roy
- Department
of Biological Sciences, Indian Institute
of Science Education and Research (IISER) Kolkata, Mohanpur741246, India
- Department
of Chemistry, Oklahoma State University, Stillwater, Oklahoma74078, United States
| | - Neelanjana Sengupta
- Department
of Biological Sciences, Indian Institute
of Science Education and Research (IISER) Kolkata, Mohanpur741246, India
| |
Collapse
|
14
|
Qing R, Hao S, Smorodina E, Jin D, Zalevsky A, Zhang S. Protein Design: From the Aspect of Water Solubility and Stability. Chem Rev 2022; 122:14085-14179. [PMID: 35921495 PMCID: PMC9523718 DOI: 10.1021/acs.chemrev.1c00757] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Indexed: 12/13/2022]
Abstract
Water solubility and structural stability are key merits for proteins defined by the primary sequence and 3D-conformation. Their manipulation represents important aspects of the protein design field that relies on the accurate placement of amino acids and molecular interactions, guided by underlying physiochemical principles. Emulated designer proteins with well-defined properties both fuel the knowledge-base for more precise computational design models and are used in various biomedical and nanotechnological applications. The continuous developments in protein science, increasing computing power, new algorithms, and characterization techniques provide sophisticated toolkits for solubility design beyond guess work. In this review, we summarize recent advances in the protein design field with respect to water solubility and structural stability. After introducing fundamental design rules, we discuss the transmembrane protein solubilization and de novo transmembrane protein design. Traditional strategies to enhance protein solubility and structural stability are introduced. The designs of stable protein complexes and high-order assemblies are covered. Computational methodologies behind these endeavors, including structure prediction programs, machine learning algorithms, and specialty software dedicated to the evaluation of protein solubility and aggregation, are discussed. The findings and opportunities for Cryo-EM are presented. This review provides an overview of significant progress and prospects in accurate protein design for solubility and stability.
Collapse
Affiliation(s)
- Rui Qing
- State
Key Laboratory of Microbial Metabolism, School of Life Sciences and
Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Media
Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- The
David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Shilei Hao
- Media
Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Key
Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Eva Smorodina
- Department
of Immunology, University of Oslo and Oslo
University Hospital, Oslo 0424, Norway
| | - David Jin
- Avalon GloboCare
Corp., Freehold, New Jersey 07728, United States
| | - Arthur Zalevsky
- Laboratory
of Bioinformatics Approaches in Combinatorial Chemistry and Biology, Shemyakin−Ovchinnikov Institute of Bioorganic
Chemistry RAS, Moscow 117997, Russia
| | - Shuguang Zhang
- Media
Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
15
|
Zhu H, Tian F, Sun L, Zhu Y, Qiu Q, Dai L. Computational Design of Extraordinarily Stable Peptide Structures through Side-Chain-Locked Knots. J Phys Chem Lett 2022; 13:7741-7748. [PMID: 35969173 DOI: 10.1021/acs.jpclett.2c02385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Extraordinarily stable protein and peptide structures are critically demanded in many applications. Typical approaches to enhance protein and peptide stability are strengthening certain interactions. Here, we develop a very different approach: stabilizing peptide structures through side-chain-locked knots. More specifically, a peptide core consists of a knot, which is prevented from unknotting and unfolding by large side chains of amino acids at knot boundaries. These side chains impose free energy barriers for unknotting. The free energy barriers are quantified using all-atom and coarse-grained simulations. The barriers become infinitely high for large side chains and tight knot cores, resulting in stable peptide structures, which never unfold unless one chemical bond is broken. The extraordinary stability is essentially kinetic stability. Our new approach lifts the thermodynamic restriction in designing peptide structures, provides extra freedom in selecting sequence and structural motifs that are thermodynamically unstable, and should expand the functionality of peptides. This work also provides a bottom-up understanding of how knotting enhances protein stability.
Collapse
Affiliation(s)
- Haoqi Zhu
- Department of Physics, City University of Hong Kong, Kowloon 999077, Hong Kong Special Administrative Region of the People's Republic of China
| | - Fujia Tian
- Department of Physics, City University of Hong Kong, Kowloon 999077, Hong Kong Special Administrative Region of the People's Republic of China
| | - Liang Sun
- Department of Physics, City University of Hong Kong, Kowloon 999077, Hong Kong Special Administrative Region of the People's Republic of China
| | - Yongjian Zhu
- Department of Physics, City University of Hong Kong, Kowloon 999077, Hong Kong Special Administrative Region of the People's Republic of China
| | - Qiyuan Qiu
- Department of Physics, City University of Hong Kong, Kowloon 999077, Hong Kong Special Administrative Region of the People's Republic of China
| | - Liang Dai
- Department of Physics, City University of Hong Kong, Kowloon 999077, Hong Kong Special Administrative Region of the People's Republic of China
| |
Collapse
|
16
|
Turzo SMBA, Seffernick JT, Rolland AD, Donor MT, Heinze S, Prell JS, Wysocki VH, Lindert S. Protein shape sampled by ion mobility mass spectrometry consistently improves protein structure prediction. Nat Commun 2022; 13:4377. [PMID: 35902583 PMCID: PMC9334640 DOI: 10.1038/s41467-022-32075-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 07/14/2022] [Indexed: 11/09/2022] Open
Abstract
Ion mobility (IM) mass spectrometry provides structural information about protein shape and size in the form of an orientationally-averaged collision cross-section (CCSIM). While IM data have been used with various computational methods, they have not yet been utilized to predict monomeric protein structure from sequence. Here, we show that IM data can significantly improve protein structure determination using the modelling suite Rosetta. We develop the Rosetta Projection Approximation using Rough Circular Shapes (PARCS) algorithm that allows for fast and accurate prediction of CCSIM from structure. Following successful testing of the PARCS algorithm, we use an integrative modelling approach to utilize IM data for protein structure prediction. Additionally, we propose a confidence metric that identifies near native models in the absence of a known structure. The results of this study demonstrate the ability of IM data to consistently improve protein structure prediction.
Collapse
Affiliation(s)
- S M Bargeen Alam Turzo
- Department of Chemistry and Biochemistry and Resource for Native Mass Spectrometry Guided Structural Biology, Ohio State University, Columbus, OH, 43210, USA
| | - Justin T Seffernick
- Department of Chemistry and Biochemistry and Resource for Native Mass Spectrometry Guided Structural Biology, Ohio State University, Columbus, OH, 43210, USA
| | - Amber D Rolland
- Department of Chemistry and Biochemistry and Materials Science Institute, University of Oregon, Eugene, OR, 97403, USA
| | - Micah T Donor
- Department of Chemistry and Biochemistry and Materials Science Institute, University of Oregon, Eugene, OR, 97403, USA
| | - Sten Heinze
- Department of Chemistry and Biochemistry and Resource for Native Mass Spectrometry Guided Structural Biology, Ohio State University, Columbus, OH, 43210, USA
| | - James S Prell
- Department of Chemistry and Biochemistry and Materials Science Institute, University of Oregon, Eugene, OR, 97403, USA
| | - Vicki H Wysocki
- Department of Chemistry and Biochemistry and Resource for Native Mass Spectrometry Guided Structural Biology, Ohio State University, Columbus, OH, 43210, USA
| | - Steffen Lindert
- Department of Chemistry and Biochemistry and Resource for Native Mass Spectrometry Guided Structural Biology, Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
17
|
Medina-Ortiz D, Contreras S, Amado-Hinojosa J, Torres-Almonacid J, Asenjo JA, Navarrete M, Olivera-Nappa Á. Generalized Property-Based Encoders and Digital Signal Processing Facilitate Predictive Tasks in Protein Engineering. Front Mol Biosci 2022; 9:898627. [PMID: 35911960 PMCID: PMC9329607 DOI: 10.3389/fmolb.2022.898627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Computational methods in protein engineering often require encoding amino acid sequences, i.e., converting them into numeric arrays. Physicochemical properties are a typical choice to define encoders, where we replace each amino acid by its value for a given property. However, what property (or group thereof) is best for a given predictive task remains an open problem. In this work, we generalize property-based encoding strategies to maximize the performance of predictive models in protein engineering. First, combining text mining and unsupervised learning, we partitioned the AAIndex database into eight semantically-consistent groups of properties. We then applied a non-linear PCA within each group to define a single encoder to represent it. Then, in several case studies, we assess the performance of predictive models for protein and peptide function, folding, and biological activity, trained using the proposed encoders and classical methods (One Hot Encoder and TAPE embeddings). Models trained on datasets encoded with our encoders and converted to signals through the Fast Fourier Transform (FFT) increased their precision and reduced their overfitting substantially, outperforming classical approaches in most cases. Finally, we propose a preliminary methodology to create de novo sequences with desired properties. All these results offer simple ways to increase the performance of general and complex predictive tasks in protein engineering without increasing their complexity.
Collapse
Affiliation(s)
- David Medina-Ortiz
- Centre for Biotechnology and Bioengineering, Universidad de Chile, Santiago, Chile
- Departamento de Ingeniería en Computación, Universidad de Magallanes, Punta Arenas, Chile
| | - Sebastian Contreras
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
- *Correspondence: Sebastian Contreras, ; Álvaro Olivera-Nappa,
| | - Juan Amado-Hinojosa
- Centre for Biotechnology and Bioengineering, Universidad de Chile, Santiago, Chile
- Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Santiago, Chile
| | - Jorge Torres-Almonacid
- Departamento de Ingeniería en Computación, Universidad de Magallanes, Punta Arenas, Chile
| | - Juan A. Asenjo
- Centre for Biotechnology and Bioengineering, Universidad de Chile, Santiago, Chile
- Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Santiago, Chile
| | | | - Álvaro Olivera-Nappa
- Centre for Biotechnology and Bioengineering, Universidad de Chile, Santiago, Chile
- Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Santiago, Chile
- *Correspondence: Sebastian Contreras, ; Álvaro Olivera-Nappa,
| |
Collapse
|
18
|
Ding W, Nakai K, Gong H. Protein design via deep learning. Brief Bioinform 2022; 23:bbac102. [PMID: 35348602 PMCID: PMC9116377 DOI: 10.1093/bib/bbac102] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/26/2022] [Accepted: 03/01/2022] [Indexed: 12/11/2022] Open
Abstract
Proteins with desired functions and properties are important in fields like nanotechnology and biomedicine. De novo protein design enables the production of previously unseen proteins from the ground up and is believed as a key point for handling real social challenges. Recent introduction of deep learning into design methods exhibits a transformative influence and is expected to represent a promising and exciting future direction. In this review, we retrospect the major aspects of current advances in deep-learning-based design procedures and illustrate their novelty in comparison with conventional knowledge-based approaches through noticeable cases. We not only describe deep learning developments in structure-based protein design and direct sequence design, but also highlight recent applications of deep reinforcement learning in protein design. The future perspectives on design goals, challenges and opportunities are also comprehensively discussed.
Collapse
Affiliation(s)
- Wenze Ding
- School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing 210044, China
- School of Future Technology, Nanjing University of Information Science and Technology, Nanjing 210044, China
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China
| | - Kenta Nakai
- Institute of Medical Science, the University of Tokyo, Tokyo 1088639, Japan
| | - Haipeng Gong
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
19
|
Boral A, Khamaru M, Mitra D. Designing synthetic transcription factors: A structural perspective. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 130:245-287. [PMID: 35534109 DOI: 10.1016/bs.apcsb.2021.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In this chapter, we discuss different design strategies of synthetic proteins, especially synthetic transcription factors. Design and engineering of synthetic transcription factors is particularly relevant for the need-based manipulation of gene expression. With recent advances in structural biology techniques and with the emergence of other precision biochemical/physical tools, accurate knowledge on structure-function relations is increasingly becoming available. Besides discussing the underlying principles of design, we go through individual cases, especially those involving four major groups of transcription factors-basic leucine zippers, zinc fingers, helix-turn-helix and homeodomains. We further discuss how synthetic biology can come together with structural biology to alter the genetic blueprint of life.
Collapse
Affiliation(s)
- Aparna Boral
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| | - Madhurima Khamaru
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| | - Devrani Mitra
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India.
| |
Collapse
|
20
|
Dwivedi KA, Huang SJ, Wang CT. Integration of various technology-based approaches for enhancing the performance of microbial fuel cell technology: A review. CHEMOSPHERE 2022; 287:132248. [PMID: 34543899 DOI: 10.1016/j.chemosphere.2021.132248] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/14/2021] [Accepted: 09/12/2021] [Indexed: 06/13/2023]
Abstract
The conflict between climate change and growing global energy demand is an immense sustainability challenge that requires noteworthy scientific and technological developments. Recently the importance of microbial fuel cell (MFC) on this issue has seen profound investigation due to its inherent ability of simultaneous wastewater treatment, and power production. However, the challenges of economy-related manufacturing and operation costs should be lowered to achieve positive field-scale demonstration. Also, a variety of different field deployments will lead to improvisation. Hence, this review article discusses the possibility of integration of MFC technology with various technologies of recent times leading to advanced sustainable MFC technology. Technological innovation in the field of nanotechnology, genetic engineering, additive manufacturing, artificial intelligence, adaptive control, and few other hybrid systems integrated with MFCs is discussed. This comprehensive and state-of-the-art study elaborates hybrid MFCs integrated with various technology and its working principles, modified electrode material, complex and easy to manufacture reactor designs, and the effects of various operating parameters on system performances. Although integrated systems are promising, much future research work is needed to overcome the challenges and commercialize hybrid MFC technology.
Collapse
Affiliation(s)
- Kavya Arun Dwivedi
- Department of Mechanical Engineering, National Taiwan University of Science and Technology, No. 43, Section 4, Keelung Road, Taipei, Taiwan
| | - Song-Jeng Huang
- Department of Mechanical Engineering, National Taiwan University of Science and Technology, No. 43, Section 4, Keelung Road, Taipei, Taiwan
| | - Chin-Tsan Wang
- Department of Mechanical and Electromechanical Engineering, National I Lan University, I Lan, Taiwan; Department of Chemical Engineering, Indian Institute of Technology Guwahati, Assam, India.
| |
Collapse
|
21
|
Sinha NJ, Langenstein MG, Pochan DJ, Kloxin CJ, Saven JG. Peptide Design and Self-assembly into Targeted Nanostructure and Functional Materials. Chem Rev 2021; 121:13915-13935. [PMID: 34709798 DOI: 10.1021/acs.chemrev.1c00712] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Peptides have been extensively utilized to construct nanomaterials that display targeted structure through hierarchical assembly. The self-assembly of both rationally designed peptides derived from naturally occurring domains in proteins as well as intuitively or computationally designed peptides that form β-sheets and helical secondary structures have been widely successful in constructing nanoscale morphologies with well-defined 1-d, 2-d, and 3-d architectures. In this review, we discuss these successes of peptide self-assembly, especially in the context of designing hierarchical materials. In particular, we emphasize the differences in the level of peptide design as an indicator of complexity within the targeted self-assembled materials and highlight future avenues for scientific and technological advances in this field.
Collapse
Affiliation(s)
- Nairiti J Sinha
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Matthew G Langenstein
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Darrin J Pochan
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Christopher J Kloxin
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States.,Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Jeffery G Saven
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
22
|
López-Arvizu A, Rocha-Mendoza D, Farrés A, Ponce-Alquicira E, García-Cano I. Improved antimicrobial spectrum of the N-acetylmuramoyl-L-alanine amidase from Latilactobacillus sakei upon LysM domain deletion. World J Microbiol Biotechnol 2021; 37:196. [PMID: 34654973 DOI: 10.1007/s11274-021-03169-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022]
Abstract
The gene encoding N-acetylmuramoyl-L-alanine amidase in Latilactobacillus sakei isolated from a fermented meat product was cloned in two forms: its complete sequence (AmiC) and a truncated sequence without one of its anchoring LysM domains (AmiLysM4). The objective of this work was to evaluate the effect of LysM domain deletion on antibacterial activity as well the biochemical characterization of each recombinant protein. AmiC and AmiLysM4 were expressed in Escherichia coli BL21. Using a zymography method, two bands with lytic activity were observed, which were confirmed by LC-MS/MS analysis, with molecular masses of 71 kDa (AmiC) and 66 kDa (AmiLysM4). The recombinant proteins were active against Listeria innocua and Staphylococcus aureus strains. The inhibitory spectrum of AmiLysM4 was broader than AmiC as it showed inhibition of Leuconostoc mesenteroides and Weissella viridescens, both microorganisms associated with food decomposition. Optimal temperature and pH values were determined for both proteins using L-alanine-p-nitroanilide hydrochloride as a substrate for N-acetylmuramoyl-L-alanine amidase activity. Both proteins showed similar maximum activity values for pH (8) and temperature (50 °C). Furthermore, structural predictions did not show differences for the catalytic region, but differences were found for the region called 2dom-AmiLysM4, which includes 4 of the 5 LysM domains. Therefore, modification of the LysM domain offers new tools for the development of novel food biopreservatives.
Collapse
Affiliation(s)
- Adriana López-Arvizu
- Departamento de Biotecnología, Universidad Autónoma Metropolitana Unidad Iztapalapa, Mexico, México
| | - Diana Rocha-Mendoza
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, USA
| | - Amelia Farrés
- Departamento de Alimentos y Biotecnología, Facultad de Química UNAM, Mexico, México
| | - Edith Ponce-Alquicira
- Departamento de Biotecnología, Universidad Autónoma Metropolitana Unidad Iztapalapa, Mexico, México.
| | - Israel García-Cano
- Departamento de Biotecnología, Universidad Autónoma Metropolitana Unidad Iztapalapa, Mexico, México. .,Department of Food Science and Technology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
23
|
Saikia B, Gogoi CR, Rahman A, Baruah A. Identification of an optimal foldability criterion to design misfolding resistant protein. J Chem Phys 2021; 155:144102. [PMID: 34654294 DOI: 10.1063/5.0057533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Proteins achieve their functional, active, and operative three dimensional native structures by overcoming the possibility of being trapped in non-native energy minima present in the energy landscape. The enormous and intricate interactions that play an important role in protein folding also determine the stability of the proteins. The large number of stabilizing/destabilizing interactions makes proteins to be only marginally stable as compared to the other competing structures. Therefore, there are some possibilities that they become trapped in the non-native conformations and thus get misfolded. These misfolded proteins lead to several debilitating diseases. This work performs a comparative study of some existing foldability criteria in the computational design of misfold resistant protein sequences based on self-consistent mean field theory. The foldability criteria selected for this study are Ef, Δ, and Φ that are commonly used in protein design procedures to determine the most efficient foldability criterion for the design of misfolding resistant proteins. The results suggest that the foldability criterion Δ is significantly better in designing a funnel energy landscape stabilizing the target state. The results also suggest that inclusion of negative design features is important for designing misfolding resistant proteins, but more information about the non-native conformations in terms of Φ leads to worse results compared to even simple positive design. The sequences designed using Δ show better resistance to misfolding in the Monte Carlo simulations performed in the study.
Collapse
Affiliation(s)
- Bondeepa Saikia
- Department of Chemistry, Dibrugarh University, Dibrugarh 786004, India
| | - Chimi Rekha Gogoi
- Department of Chemistry, Dibrugarh University, Dibrugarh 786004, India
| | - Aziza Rahman
- Department of Chemistry, Dibrugarh University, Dibrugarh 786004, India
| | - Anupaul Baruah
- Department of Chemistry, Dibrugarh University, Dibrugarh 786004, India
| |
Collapse
|
24
|
Arnittali M, Rissanou AN, Amprazi M, Kokkinidis M, Harmandaris V. Structure and Thermal Stability of wtRop and RM6 Proteins through All-Atom Molecular Dynamics Simulations and Experiments. Int J Mol Sci 2021; 22:ijms22115931. [PMID: 34073028 PMCID: PMC8199364 DOI: 10.3390/ijms22115931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 01/07/2023] Open
Abstract
In the current work we study, via molecular simulations and experiments, the folding and stability of proteins from the tertiary motif of 4-α-helical bundles, a recurrent motif consisting of four amphipathic α-helices packed in a parallel or antiparallel fashion. The focus is on the role of the loop region in the structure and the properties of the wild-type Rop (wtRop) and RM6 proteins, exploring the key factors which can affect them, through all-atom molecular dynamics (MD) simulations and supporting by experimental findings. A detailed investigation of structural and conformational properties of wtRop and its RM6 loopless mutation is presented, which display different physical characteristics even in their native states. Then, the thermal stability of both proteins is explored showing RM6 as more thermostable than wtRop through all studied measures. Deviations from native structures are detected mostly in tails and loop regions and most flexible residues are indicated. Decrease of hydrogen bonds with the increase of temperature is observed, as well as reduction of hydrophobic contacts in both proteins. Experimental data from circular dichroism spectroscopy (CD), are also presented, highlighting the effect of temperature on the structural integrity of wtRop and RM6. The central goal of this study is to explore on the atomic level how a protein mutation can cause major changes in its physical properties, like its structural stability.
Collapse
Affiliation(s)
- Maria Arnittali
- Institute of Applied and Computational Mathematics (IACM), Foundation for Research and Technology Hellas (FORTH), IACM/FORTH, GR-71110 Heraklion, Crete, Greece; (M.A.); (V.H.)
- Department of Mathematics and Applied Mathematics, University of Crete, GR-71409 Heraklion, Crete, Greece
| | - Anastassia N. Rissanou
- Institute of Applied and Computational Mathematics (IACM), Foundation for Research and Technology Hellas (FORTH), IACM/FORTH, GR-71110 Heraklion, Crete, Greece; (M.A.); (V.H.)
- Department of Mathematics and Applied Mathematics, University of Crete, GR-71409 Heraklion, Crete, Greece
- Correspondence: ; Tel.: +30-2810-393746
| | - Maria Amprazi
- Department of Biology, University of Crete, GR-71409 Heraklion, Crete, Greece; (M.A.); (M.K.)
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, GR-70013 Heraklion, Crete, Greece
| | - Michael Kokkinidis
- Department of Biology, University of Crete, GR-71409 Heraklion, Crete, Greece; (M.A.); (M.K.)
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, GR-70013 Heraklion, Crete, Greece
| | - Vagelis Harmandaris
- Institute of Applied and Computational Mathematics (IACM), Foundation for Research and Technology Hellas (FORTH), IACM/FORTH, GR-71110 Heraklion, Crete, Greece; (M.A.); (V.H.)
- Department of Mathematics and Applied Mathematics, University of Crete, GR-71409 Heraklion, Crete, Greece
- Computation-Based Science and Technology Research Center, The Cyprus Institute, 2121 Nicosia, Cyprus
| |
Collapse
|
25
|
Subramanian RH, Suzuki Y, Tallorin L, Sahu S, Thompson M, Gianneschi NC, Burkart MD, Tezcan FA. Enzyme-Directed Functionalization of Designed, Two-Dimensional Protein Lattices. Biochemistry 2021; 60:1050-1062. [PMID: 32706243 PMCID: PMC7855359 DOI: 10.1021/acs.biochem.0c00363] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The design and construction of crystalline protein arrays to selectively assemble ordered nanoscale materials have potential applications in sensing, catalysis, and medicine. Whereas numerous designs have been implemented for the bottom-up construction of protein assemblies, the generation of artificial functional materials has been relatively unexplored. Enzyme-directed post-translational modifications are responsible for the functional diversity of the proteome and, thus, could be harnessed to selectively modify artificial protein assemblies. In this study, we describe the use of phosphopantetheinyl transferases (PPTases), a class of enzymes that covalently modify proteins using coenzyme A (CoA), to site-selectively tailor the surface of designed, two-dimensional (2D) protein crystals. We demonstrate that a short peptide (ybbR) or a molecular tag (CoA) can be covalently tethered to 2D arrays to enable enzymatic functionalization using Sfp PPTase. The site-specific modification of two different protein array platforms is facilitated by PPTases to afford both small molecule- and protein-functionalized surfaces with no loss of crystalline order. This work highlights the potential for chemoenzymatic modification of large protein surfaces toward the generation of sophisticated protein platforms reminiscent of the complex landscape of cell surfaces.
Collapse
Affiliation(s)
- Rohit H. Subramanian
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
| | - Yuta Suzuki
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
- Current address: Hakubi Center for Advanced Research, Kyoto University, Yoshida-honmachi, Sakyo-ku, Kyoto, Japan, 606-8501
| | - Lorillee Tallorin
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
| | - Swagat Sahu
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
| | - Matthew Thompson
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
- Departments of Chemistry, Materials Science & Engineering, Biomedical Engineering, Chemistry of Life Processes Institute, International Institute for Nanotechnology, Simpson Querrey Institute, Northwestern University, Evanston, Illinois 60208, USA
| | - Nathan C. Gianneschi
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
- Departments of Chemistry, Materials Science & Engineering, Biomedical Engineering, Chemistry of Life Processes Institute, International Institute for Nanotechnology, Simpson Querrey Institute, Northwestern University, Evanston, Illinois 60208, USA
| | - Michael D. Burkart
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
| | - F. Akif Tezcan
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
- Materials Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
26
|
de Araújo RSA, Mendonça FJ, Scotti MT, Scotti L. Protein modeling. PHYSICAL SCIENCES REVIEWS 2021. [DOI: 10.1515/psr-2018-0161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Proteins are essential and versatile polymers consisting of sequenced amino acids that often possess an organized three-dimensional arrangement, (a result of their monomeric composition), which determines their biological role in cellular function. Proteins are involved in enzymatic catalysis; they participate in genetic information decoding and transmission processes, in cell recognition, in signaling, and transport of substances, in regulation of intra and extracellular conditions, and other functions.
Collapse
Affiliation(s)
- Rodrigo S. A. de Araújo
- Biological Science Department, Laboratory of Synthesis and Drug Delivery , State University of Paraiba , 58070-450 , João Pessoa , PB , Brazil
| | - Francisco J. B. Mendonça
- Biological Science Department, Laboratory of Synthesis and Drug Delivery , State University of Paraiba , 58070-450 , João Pessoa , PB , Brazil
| | - Marcus T. Scotti
- Health Center , Federal University of Paraíba , 50670-910 , João Pessoa , PB , Brazil
| | - Luciana Scotti
- Health Center , Federal University of Paraíba , 50670-910 , João Pessoa , PB , Brazil
| |
Collapse
|
27
|
Kefala A, Amprazi M, Mylonas E, Kotsifaki D, Providaki M, Pozidis C, Fotiadou M, Kokkinidis M. Probing Protein Folding with Sequence-Reversed α-Helical Bundles. Int J Mol Sci 2021; 22:ijms22041955. [PMID: 33669383 PMCID: PMC7920257 DOI: 10.3390/ijms22041955] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/11/2021] [Accepted: 02/11/2021] [Indexed: 12/22/2022] Open
Abstract
Recurrent protein folding motifs include various types of helical bundles formed by α-helices that supercoil around each other. While specific patterns of amino acid residues (heptad repeats) characterize the highly versatile folding motif of four-α-helical bundles, the significance of the polypeptide chain directionality is not sufficiently understood, although it determines sequence patterns, helical dipoles, and other parameters for the folding and oligomerization processes of bundles. To investigate directionality aspects in sequence-structure relationships, we reversed the amino acid sequences of two well-characterized, highly regular four-α-helical bundle proteins and studied the folding, oligomerization, and structural properties of the retro-proteins, using Circular Dichroism Spectroscopy (CD), Size Exclusion Chromatography combined with Multi-Angle Laser Light Scattering (SEC-MALS), and Small Angle X-ray Scattering (SAXS). The comparison of the parent proteins with their retro-counterparts reveals that while the α-helical character of the parents is affected to varying degrees by sequence reversal, the folding states, oligomerization propensities, structural stabilities, and shapes of the new molecules strongly depend on the characteristics of the heptad repeat patterns. The highest similarities between parent and retro-proteins are associated with the presence of uninterrupted heptad patterns in helical bundles sequences.
Collapse
Affiliation(s)
- Aikaterini Kefala
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology–Hellas (IMBB-FORTH), 70013 Heraklion, Greece; (A.K.); (M.A.); (E.M.); (D.K.); (M.P.); (C.P.)
- Department of Biology, University of Crete, 70013 Heraklion, Greece;
| | - Maria Amprazi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology–Hellas (IMBB-FORTH), 70013 Heraklion, Greece; (A.K.); (M.A.); (E.M.); (D.K.); (M.P.); (C.P.)
- Department of Biology, University of Crete, 70013 Heraklion, Greece;
| | - Efstratios Mylonas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology–Hellas (IMBB-FORTH), 70013 Heraklion, Greece; (A.K.); (M.A.); (E.M.); (D.K.); (M.P.); (C.P.)
| | - Dina Kotsifaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology–Hellas (IMBB-FORTH), 70013 Heraklion, Greece; (A.K.); (M.A.); (E.M.); (D.K.); (M.P.); (C.P.)
| | - Mary Providaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology–Hellas (IMBB-FORTH), 70013 Heraklion, Greece; (A.K.); (M.A.); (E.M.); (D.K.); (M.P.); (C.P.)
| | - Charalambos Pozidis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology–Hellas (IMBB-FORTH), 70013 Heraklion, Greece; (A.K.); (M.A.); (E.M.); (D.K.); (M.P.); (C.P.)
| | - Melina Fotiadou
- Department of Biology, University of Crete, 70013 Heraklion, Greece;
| | - Michael Kokkinidis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology–Hellas (IMBB-FORTH), 70013 Heraklion, Greece; (A.K.); (M.A.); (E.M.); (D.K.); (M.P.); (C.P.)
- Department of Biology, University of Crete, 70013 Heraklion, Greece;
- Correspondence: ; Tel.: +30-2810-394350
| |
Collapse
|
28
|
Pham PN, Huličiak M, Biedermannová L, Černý J, Charnavets T, Fuertes G, Herynek Š, Kolářová L, Kolenko P, Pavlíček J, Zahradník J, Mikulecky P, Schneider B. Protein Binder (ProBi) as a New Class of Structurally Robust Non-Antibody Protein Scaffold for Directed Evolution. Viruses 2021; 13:v13020190. [PMID: 33514045 PMCID: PMC7911045 DOI: 10.3390/v13020190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/15/2021] [Accepted: 01/23/2021] [Indexed: 12/13/2022] Open
Abstract
Engineered small non-antibody protein scaffolds are a promising alternative to antibodies and are especially attractive for use in protein therapeutics and diagnostics. The advantages include smaller size and a more robust, single-domain structural framework with a defined binding surface amenable to mutation. This calls for a more systematic approach in designing new scaffolds suitable for use in one or more methods of directed evolution. We hereby describe a process based on an analysis of protein structures from the Protein Data Bank and their experimental examination. The candidate protein scaffolds were subjected to a thorough screening including computational evaluation of the mutability, and experimental determination of their expression yield in E. coli, solubility, and thermostability. In the next step, we examined several variants of the candidate scaffolds including their wild types and alanine mutants. We proved the applicability of this systematic procedure by selecting a monomeric single-domain human protein with a fold different from previously known scaffolds. The newly developed scaffold, called ProBi (Protein Binder), contains two independently mutable surface patches. We demonstrated its functionality by training it as a binder against human interleukin-10, a medically important cytokine. The procedure yielded scaffold-related variants with nanomolar affinity.
Collapse
|
29
|
Arai K, Iwaoka M. Flexible Folding: Disulfide-Containing Peptides and Proteins Choose the Pathway Depending on the Environments. Molecules 2021; 26:E195. [PMID: 33401729 PMCID: PMC7794709 DOI: 10.3390/molecules26010195] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 12/24/2020] [Accepted: 12/29/2020] [Indexed: 11/18/2022] Open
Abstract
In the last few decades, development of novel experimental techniques, such as new types of disulfide (SS)-forming reagents and genetic and chemical technologies for synthesizing designed artificial proteins, is opening a new realm of the oxidative folding study where peptides and proteins can be folded under physiologically more relevant conditions. In this review, after a brief overview of the historical and physicochemical background of oxidative protein folding study, recently revealed folding pathways of several representative peptides and proteins are summarized, including those having two, three, or four SS bonds in the native state, as well as those with odd Cys residues or consisting of two peptide chains. Comparison of the updated pathways with those reported in the early years has revealed the flexible nature of the protein folding pathways. The significantly different pathways characterized for hen-egg white lysozyme and bovine milk α-lactalbumin, which belong to the same protein superfamily, suggest that the information of protein folding pathways, not only the native folded structure, is encoded in the amino acid sequence. The application of the flexible pathways of peptides and proteins to the engineering of folded three-dimensional structures is an interesting and important issue in the new realm of the current oxidative protein folding study.
Collapse
Affiliation(s)
| | - Michio Iwaoka
- Department of Chemistry, School of Science, Tokai University, Kitakaname, Hiratsuka-shi, Kanagawa 259-1292, Japan;
| |
Collapse
|
30
|
Abstract
This chapter describes two computational methods for PDZ-peptide binding: high-throughput computational protein design (CPD) and a medium-throughput approach combining molecular dynamics for conformational sampling with a Poisson-Boltzmann (PB) Linear Interaction Energy for scoring. A new CPD method is outlined, which uses adaptive Monte Carlo simulations to efficiently sample peptide variants that tightly bind a PDZ domain, and provides at the same time precise estimates of their relative binding free energies. A detailed protocol is described based on the Proteus CPD software. The medium-throughput approach can be performed with standard MD and PB software, such as NAMD and Charmm. For 40 complexes between Tiam1 and peptide ligands, it gave high a2ccuracy, with mean errors of around 0.5 kcal/mol for relative binding free energies and no large errors. It requires a moderate amount of parameter fitting before it can be applied, and its transferability to other protein families is still untested.
Collapse
Affiliation(s)
- Nicolas Panel
- Laboratoire de Biologie Structurale de la Cellule (CNRS UMR7654), Ecole Polytechnique, Palaiseau, France
| | - Francesco Villa
- Laboratoire de Biologie Structurale de la Cellule (CNRS UMR7654), Ecole Polytechnique, Palaiseau, France
| | - Vaitea Opuu
- Laboratoire de Biologie Structurale de la Cellule (CNRS UMR7654), Ecole Polytechnique, Palaiseau, France
| | - David Mignon
- Laboratoire de Biologie Structurale de la Cellule (CNRS UMR7654), Ecole Polytechnique, Palaiseau, France
| | - Thomas Simonson
- Laboratoire de Biologie Structurale de la Cellule (CNRS UMR7654), Ecole Polytechnique, Palaiseau, France.
| |
Collapse
|
31
|
Unger EK, Keller JP, Altermatt M, Liang R, Matsui A, Dong C, Hon OJ, Yao Z, Sun J, Banala S, Flanigan ME, Jaffe DA, Hartanto S, Carlen J, Mizuno GO, Borden PM, Shivange AV, Cameron LP, Sinning S, Underhill SM, Olson DE, Amara SG, Temple Lang D, Rudnick G, Marvin JS, Lavis LD, Lester HA, Alvarez VA, Fisher AJ, Prescher JA, Kash TL, Yarov-Yarovoy V, Gradinaru V, Looger LL, Tian L. Directed Evolution of a Selective and Sensitive Serotonin Sensor via Machine Learning. Cell 2020; 183:1986-2002.e26. [PMID: 33333022 PMCID: PMC8025677 DOI: 10.1016/j.cell.2020.11.040] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 06/22/2020] [Accepted: 11/20/2020] [Indexed: 12/28/2022]
Abstract
Serotonin plays a central role in cognition and is the target of most pharmaceuticals for psychiatric disorders. Existing drugs have limited efficacy; creation of improved versions will require better understanding of serotonergic circuitry, which has been hampered by our inability to monitor serotonin release and transport with high spatial and temporal resolution. We developed and applied a binding-pocket redesign strategy, guided by machine learning, to create a high-performance, soluble, fluorescent serotonin sensor (iSeroSnFR), enabling optical detection of millisecond-scale serotonin transients. We demonstrate that iSeroSnFR can be used to detect serotonin release in freely behaving mice during fear conditioning, social interaction, and sleep/wake transitions. We also developed a robust assay of serotonin transporter function and modulation by drugs. We expect that both machine-learning-guided binding-pocket redesign and iSeroSnFR will have broad utility for the development of other sensors and in vitro and in vivo serotonin detection, respectively.
Collapse
Affiliation(s)
- Elizabeth K Unger
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Jacob P Keller
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20174, USA
| | - Michael Altermatt
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ruqiang Liang
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Aya Matsui
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Chunyang Dong
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Olivia J Hon
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Zi Yao
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Junqing Sun
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Samba Banala
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20174, USA
| | - Meghan E Flanigan
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - David A Jaffe
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Samantha Hartanto
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Jane Carlen
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Grace O Mizuno
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Phillip M Borden
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20174, USA
| | - Amol V Shivange
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Lindsay P Cameron
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Steffen Sinning
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Suzanne M Underhill
- Laboratory of Molecular and Cellular Neurobiology, National Institute on Mental Health, NIH, Bethesda, MD 20892, USA
| | - David E Olson
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Susan G Amara
- Laboratory of Molecular and Cellular Neurobiology, National Institute on Mental Health, NIH, Bethesda, MD 20892, USA
| | - Duncan Temple Lang
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Gary Rudnick
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jonathan S Marvin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20174, USA
| | - Luke D Lavis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20174, USA
| | - Henry A Lester
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Andrew J Fisher
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Jennifer A Prescher
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Vladimir Yarov-Yarovoy
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Loren L Looger
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20174, USA.
| | - Lin Tian
- Departments of Biochemistry and Molecular Medicine, Chemistry, Statistics, Molecular and Cellular Biology, and Physiology and Membrane Biology, the Center for Neuroscience, and Graduate Programs in Molecular, Cellular, and Integrative Physiology, Biochemistry, Molecular, Cellular and Developmental Biology and Neuroscience, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
32
|
Patra P, Bhattacharya M, Sharma AR, Ghosh P, Sharma G, Patra BC, Mallick B, Lee SS, Chakraborty C. Identification and Design of a Next-Generation Multi Epitopes Bases Peptide Vaccine Candidate Against Prostate Cancer: An In Silico Approach. Cell Biochem Biophys 2020; 78:495-509. [PMID: 32347457 DOI: 10.1007/s12013-020-00912-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/07/2020] [Indexed: 12/28/2022]
Abstract
Prostate cancer (PCa) is the second most diagnosed cancer in men and ranked fifth in overall cancer diagnosis. During the past decades, it has arisen as a significant life-threatening disease in men at an older age. At the early onset of illness when it is in localized form, radiation and surgical treatments are applied against this disease. In case of adverse situations androgen deprivation therapy, chemotherapy, hormonal therapy, etc. are widely used as a therapeutic element. However, studies found the occurrences of several side effects after applying these therapies. In current work, several immunoinformatic techniques were applied to formulate a multi-epitopic vaccine from the overexpressed antigenic proteins of PCa. A total of 13 epitopes were identified from the five prostatic antigenic proteins (PSA, PSMA, PSCA, STEAP, and PAP), after validation with several in silico tools. These epitopes were fused to form a vaccine element by (GGGGS)3 peptide linker. Afterward, 5, 6-dimethylxanthenone-4-acetic acid (DMXAA) was used as an adjuvant to initiate and induce STING-mediated cytotoxic cascade. In addition, molecular docking was performed between the vaccine element and HLA class I antigen with the low ACE value of -251 kcal/mol which showed a significant binding. Molecular simulation using normal mode analysis (NMA) illustrated the docking complex as a stable one. Therefore, this observation strongly indicated that our multi epitopes bases peptide vaccine molecule will be an effective candidate for the treatment of the PCa.
Collapse
Affiliation(s)
- Prasanta Patra
- Department of Zoology, Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Manojit Bhattacharya
- Department of Zoology, Vidyasagar University, Midnapore, West Bengal, 721102, India
- Institute for Skeletal Aging & Orthopedic Surgery, Chuncheon Sacred Heart Hospital, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Chuncheon Sacred Heart Hospital, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Pratik Ghosh
- Department of Zoology, Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Garima Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Bidhan Chandra Patra
- Department of Zoology, Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Bidyut Mallick
- Departments of Applied Science, Galgotias College of Engineering and Technology, Greater Noida, India
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Chuncheon Sacred Heart Hospital, Hallym University, Chuncheon, 24252, Republic of Korea.
| | - Chiranjib Chakraborty
- Institute for Skeletal Aging & Orthopedic Surgery, Chuncheon Sacred Heart Hospital, Hallym University, Chuncheon, 24252, Republic of Korea.
- Adamas University, North, 24 Parganas, Kolkata, West Bengal, 700126, India.
| |
Collapse
|
33
|
Helmy M, Smith D, Selvarajoo K. Systems biology approaches integrated with artificial intelligence for optimized metabolic engineering. Metab Eng Commun 2020; 11:e00149. [PMID: 33072513 PMCID: PMC7546651 DOI: 10.1016/j.mec.2020.e00149] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/01/2020] [Accepted: 10/07/2020] [Indexed: 12/05/2022] Open
Abstract
Metabolic engineering aims to maximize the production of bio-economically important substances (compounds, enzymes, or other proteins) through the optimization of the genetics, cellular processes and growth conditions of microorganisms. This requires detailed understanding of underlying metabolic pathways involved in the production of the targeted substances, and how the cellular processes or growth conditions are regulated by the engineering. To achieve this goal, a large system of experimental techniques, compound libraries, computational methods and data resources, including multi-omics data, are used. The recent advent of multi-omics systems biology approaches significantly impacted the field by opening new avenues to perform dynamic and large-scale analyses that deepen our knowledge on the manipulations. However, with the enormous transcriptomics, proteomics and metabolomics available, it is a daunting task to integrate the data for a more holistic understanding. Novel data mining and analytics approaches, including Artificial Intelligence (AI), can provide breakthroughs where traditional low-throughput experiment-alone methods cannot easily achieve. Here, we review the latest attempts of combining systems biology and AI in metabolic engineering research, and highlight how this alliance can help overcome the current challenges facing industrial biotechnology, especially for food-related substances and compounds using microorganisms.
Collapse
Affiliation(s)
- Mohamed Helmy
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Derek Smith
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Kumar Selvarajoo
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
- Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore (NUS), Singapore, Singapore
| |
Collapse
|
34
|
Walker SP, Yallapragada VVB, Tangney M. Arming Yourself for The In Silico Protein Design Revolution. Trends Biotechnol 2020; 39:651-664. [PMID: 33139074 DOI: 10.1016/j.tibtech.2020.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 12/23/2022]
Abstract
Proteins mediate many essential processes of life to a degree of functional precision unmatched by any synthetic device. While engineered proteins are currently used in biotech, food, biomedicine, and material technology-based industries, the true potential of proteins is practically untapped. The emerging field of in silico protein design is predicted to provide the next quantum leap in the biotech industry. Having predictive control over protein function and the ability to redefine these functions have driven the field of protein engineering into an era of unprecedented development. This article provides a holistic analysis of protein design R&D (current state-of-the-art tools and knowhow) and commercial landscape, as well as a one-stop-shop profile of in silico protein design technology for biotechnology stakeholders.
Collapse
Affiliation(s)
- Sidney P Walker
- CancerResearch@UCC, University College Cork, Cork, Ireland; SynBioCentre, University College Cork, Cork, Ireland
| | - Venkata V B Yallapragada
- CancerResearch@UCC, University College Cork, Cork, Ireland; SynBioCentre, University College Cork, Cork, Ireland
| | - Mark Tangney
- CancerResearch@UCC, University College Cork, Cork, Ireland; SynBioCentre, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
35
|
Li Q, Bu J, Ma Y, Yang J, Hu Z, Lai C, Xu Y, Tang J, Cui G, Wang Y, Zhao Y, Jin B, Shen Y, Guo J, Huang L. Characterization of O-methyltransferases involved in the biosynthesis of tetrandrine in Stephania tetrandra. JOURNAL OF PLANT PHYSIOLOGY 2020; 250:153181. [PMID: 32460036 DOI: 10.1016/j.jplph.2020.153181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 06/11/2023]
Abstract
Tetrandrine is the most effective small molecule that has been found to inhibit the Ebola virus. It is a typical bisbenzylisoquinoline alkaloid and is the main active ingredient in Stephania tetrandra. Metabolic engineering and synthetic biology are potential methods for efficient and rapid acquisition of tetrandrine. S-adenosyl-L-methionine: (S)-norcoclaurine-6-O-methyltransferase (6OMT) is a rate-limiting step involved in the biosynthesis of tetrandrine. In this study, we identify S-adenosyl-L-methionine: (S)-norcoclaurine-6-O-methyltransferase from S. tetrandra, which catalyzes the conversion of (S)-norcoclaurine to (S)-coclaurine. Four 6OMT-like genes were cloned from S. tetrandra. An in vitro enzyme assay showed that St6OMT1 could catalyze the conversion of (S)-norcoclaurine to produce (S)-coclaurine. St6OMT2 can catalyze the production of very few (S)-coclaurine molecules, accompanied by more by-products with m/z 300, compared to St6OMT1. The newly discovered 6OMTs will provide an optional genetic component for benzylisoquinoline alkaloid (BIA) synthetic biology research. This work will lay the foundation for the analysis of the biosynthetic pathway of tetrandrine in S. tetrandra.
Collapse
Affiliation(s)
- Qishuang Li
- School of Pharmacy, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, China; State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| | - Junling Bu
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| | - Ying Ma
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| | - Jian Yang
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| | - Zhimin Hu
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| | - Changjiangsheng Lai
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| | - Yanqin Xu
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, No. 1688 Meilin Avenue, Nanchang 330004, China.
| | - Jinfu Tang
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| | - Guanghong Cui
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| | - Yanan Wang
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| | - Yujun Zhao
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| | - Baolong Jin
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| | - Ye Shen
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| | - Juan Guo
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| | - Luqi Huang
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 South Side Street, Dongzhimen, Beijing 100700, China.
| |
Collapse
|
36
|
Rojas G, Orellana I, Rosales-Rojas R, García-Olivares J, Comer J, Vergara-Jaque A. Structural Determinants of the Dopamine Transporter Regulation Mediated by G Proteins. J Chem Inf Model 2020; 60:3577-3586. [PMID: 32525311 DOI: 10.1021/acs.jcim.0c00236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Dopamine clearance in the brain is controlled by the dopamine transporter (DAT), a protein residing in the plasma membrane, which drives reuptake of extracellular dopamine into presynaptic neurons. Studies have revealed that the βγ subunits of heterotrimeric G proteins modulate DAT function through a physical association with the C-terminal region of the transporter. Regulation of neurotransmitter transporters by Gβγ subunits is unprecedented in the literature; therefore, it is interesting to investigate the structural details of this particular protein-protein interaction. Here, we refined the crystal structure of the Drosophila melanogaster DAT (dDAT), modeling de novo the N- and C-terminal domains; subsequently, we used the full-length dDAT structure to generate a comparative model of human DAT (hDAT). Both proteins were assembled with Gβ1γ2 subunits employing protein-protein docking, and subsequent molecular dynamics simulations were run to identify the specific interactions governing the formation of the hDAT:Gβγ and dDAT:Gβγ complexes. A [L/F]R[Q/E]R sequence motif containing the residues R588 in hDAT and R587 in dDAT was found as key to bind the Gβγ subunits through electrostatic interactions with a cluster of negatively charged residues located at the top face of the Gβ subunit. Alterations of DAT function have been associated with multiple devastating neuropathological conditions; therefore, this work represents a step toward better understanding DAT regulation by signaling proteins, allowing us to predict therapeutic target regions.
Collapse
Affiliation(s)
- Genoveva Rojas
- Center for Bioinformatics and Molecular Simulation, Faculty of Engineering, Universidad de Talca, 2 Norte 685, Talca, Chile
| | - Ivana Orellana
- Center for Bioinformatics and Molecular Simulation, Faculty of Engineering, Universidad de Talca, 2 Norte 685, Talca, Chile
| | - Roberto Rosales-Rojas
- Center for Bioinformatics and Molecular Simulation, Faculty of Engineering, Universidad de Talca, 2 Norte 685, Talca, Chile
| | - Jennie García-Olivares
- Supernus Pharmaceuticals, 9715 Key West Avenue, Rockville, Maryland 20850, United States
| | - Jeffrey Comer
- Institute of Computational Comparative Medicine, Nanotechnology Innovation Center of Kansas State, Kansas State University, Manhattan, Kansas 66506, United States
| | - Ariela Vergara-Jaque
- Center for Bioinformatics and Molecular Simulation, Faculty of Engineering, Universidad de Talca, 2 Norte 685, Talca, Chile.,Millennium Nucleus of Ion Channels-associated Diseases (MiNICAD), Santiago, Chile
| |
Collapse
|
37
|
Yaeger-Weiss SK, Jennaro TS, Mecha M, Becker JH, Yang H, Winkler GLW, Cavagnero S. Net Charge and Nonpolar Content Guide the Identification of Folded and Prion Proteins. Biochemistry 2020; 59:1881-1895. [DOI: 10.1021/acs.biochem.9b01114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Susanna K. Yaeger-Weiss
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Theodore S. Jennaro
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Miranda Mecha
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Jenna H. Becker
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Hanming Yang
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Gordon L. W. Winkler
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Silvia Cavagnero
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
38
|
Sidky H, Chen W, Ferguson AL. Machine learning for collective variable discovery and enhanced sampling in biomolecular simulation. Mol Phys 2020. [DOI: 10.1080/00268976.2020.1737742] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Hythem Sidky
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Wei Chen
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Andrew L. Ferguson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| |
Collapse
|
39
|
Banerjee S, Mitra D. Structural Basis of Design and Engineering for Advanced Plant Optogenetics. TRENDS IN PLANT SCIENCE 2020; 25:35-65. [PMID: 31699521 DOI: 10.1016/j.tplants.2019.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 09/12/2019] [Accepted: 10/03/2019] [Indexed: 06/10/2023]
Abstract
In optogenetics, light-sensitive proteins are specifically expressed in target cells and light is used to precisely control the activity of these proteins at high spatiotemporal resolution. Optogenetics initially used naturally occurring photoreceptors to control neural circuits, but has expanded to include carefully designed and engineered photoreceptors. Several optogenetic constructs are based on plant photoreceptors, but their application to plant systems has been limited. Here, we present perspectives on the development of plant optogenetics, considering different levels of design complexity. We discuss how general principles of light-driven signal transduction can be coupled with approaches for engineering protein folding to develop novel optogenetic tools. Finally, we explore how the use of computation, networks, circular permutation, and directed evolution could enrich optogenetics.
Collapse
Affiliation(s)
- Sudakshina Banerjee
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, India
| | - Devrani Mitra
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, India.
| |
Collapse
|
40
|
Kuhlman B, Bradley P. Advances in protein structure prediction and design. Nat Rev Mol Cell Biol 2019; 20:681-697. [PMID: 31417196 PMCID: PMC7032036 DOI: 10.1038/s41580-019-0163-x] [Citation(s) in RCA: 444] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2019] [Indexed: 12/18/2022]
Abstract
The prediction of protein three-dimensional structure from amino acid sequence has been a grand challenge problem in computational biophysics for decades, owing to its intrinsic scientific interest and also to the many potential applications for robust protein structure prediction algorithms, from genome interpretation to protein function prediction. More recently, the inverse problem - designing an amino acid sequence that will fold into a specified three-dimensional structure - has attracted growing attention as a potential route to the rational engineering of proteins with functions useful in biotechnology and medicine. Methods for the prediction and design of protein structures have advanced dramatically in the past decade. Increases in computing power and the rapid growth in protein sequence and structure databases have fuelled the development of new data-intensive and computationally demanding approaches for structure prediction. New algorithms for designing protein folds and protein-protein interfaces have been used to engineer novel high-order assemblies and to design from scratch fluorescent proteins with novel or enhanced properties, as well as signalling proteins with therapeutic potential. In this Review, we describe current approaches for protein structure prediction and design and highlight a selection of the successful applications they have enabled.
Collapse
Affiliation(s)
- Brian Kuhlman
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
| | - Philip Bradley
- Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
| |
Collapse
|
41
|
Kopeć K, Pędziwiatr M, Gront D, Sztatelman O, Sławski J, Łazicka M, Worch R, Zawada K, Makarova K, Nyk M, Grzyb J. Comparison of α-Helix and β-Sheet Structure Adaptation to a Quantum Dot Geometry: Toward the Identification of an Optimal Motif for a Protein Nanoparticle Cover. ACS OMEGA 2019; 4:13086-13099. [PMID: 31460436 PMCID: PMC6705085 DOI: 10.1021/acsomega.9b00505] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/23/2019] [Indexed: 05/31/2023]
Abstract
While quantum dots (QDs) are useful as fluorescent labels, their application in biosciences is limited due to the stability and hydrophobicity of their surface. In this study, we tested two types of proteins for use as a cover for spherical QDs, composed of cadmium selenide. Pumilio homology domain (Puf), which is mostly α-helical, and leucine-rich repeat (LRR) domain, which is rich in β-sheets, were selected to determine if there is a preference for one of these secondary structure types for nanoparticle covers. The protein sequences were optimized to improve their interaction with the surface of QDs. The solubilization of the apoproteins and their assembly with nanoparticles required the application of a detergent, which was removed in subsequent steps. Finally, only the Puf-based cover was successful enough as a QD hydrophilic cover. We showed that a single polypeptide dimer of Puf, PufPuf, can form a cover. We characterized the size and fluorescent properties of the obtained QD:protein assemblies. We showed that the secondary structure of the Puf proteins was not destroyed upon contact with the QDs. We demonstrated that these assemblies do not promote the formation of reactive oxygen species during illumination of the nanoparticles. The data represent advances in the effort to obtain a stable biocompatible cover for QDs.
Collapse
Affiliation(s)
- Katarzyna Kopeć
- Institute
of Physics, Polish Academy of Sciences, Aleja Lotników 32/46, PL02668 Warsaw, Poland
| | - Marta Pędziwiatr
- Institute
of Physics, Polish Academy of Sciences, Aleja Lotników 32/46, PL02668 Warsaw, Poland
| | - Dominik Gront
- Faculty
of Chemistry, University of Warsaw, Pasteura 1, PL02093 Warsaw, Poland
| | - Olga Sztatelman
- Institute
of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, PL02106 Warsaw, Poland
| | - Jakub Sławski
- Department
of Biophysics, Faculty of Biotechnology, University of Wrocław, F. Joliot-Curie Street 14a, PL50383 Wrocław, Poland
| | - Magdalena Łazicka
- Department
of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Miecznikowa 1, PL02096 Warsaw, Poland
| | - Remigiusz Worch
- Institute
of Physics, Polish Academy of Sciences, Aleja Lotników 32/46, PL02668 Warsaw, Poland
| | - Katarzyna Zawada
- Department
of Physical Chemistry, Faculty of Pharmacy with the Laboratory Medicine
Division, The Medical University of Warsaw, Banacha 1 Street, PL02097 Warsaw, Poland
| | - Katerina Makarova
- Department
of Physical Chemistry, Faculty of Pharmacy with the Laboratory Medicine
Division, The Medical University of Warsaw, Banacha 1 Street, PL02097 Warsaw, Poland
| | - Marcin Nyk
- Advanced
Materials Engineering and Modelling Group, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego
27, PL50370 Wrocław, Poland
| | - Joanna Grzyb
- Department
of Biophysics, Faculty of Biotechnology, University of Wrocław, F. Joliot-Curie Street 14a, PL50383 Wrocław, Poland
| |
Collapse
|
42
|
Sanyal T, Mittal J, Shell MS. A hybrid, bottom-up, structurally accurate, Go¯-like coarse-grained protein model. J Chem Phys 2019; 151:044111. [PMID: 31370551 PMCID: PMC6663515 DOI: 10.1063/1.5108761] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 06/24/2019] [Indexed: 12/21/2022] Open
Abstract
Coarse-grained (CG) protein models in the structural biology literature have improved over the years from being simple tools to understand general folding and aggregation driving forces to capturing detailed structures achieved by actual folding sequences. Here, we ask whether such models can be developed systematically from recent advances in bottom-up coarse-graining methods without relying on bioinformatic data (e.g., protein data bank statistics). We use relative entropy coarse-graining to develop a hybrid CG but Go¯-like CG peptide model, hypothesizing that the landscape of proteinlike folds is encoded by the backbone interactions, while the sidechain interactions define which of these structures globally minimizes the free energy in a unique native fold. To construct a model capable of capturing varied secondary structures, we use a new extended ensemble relative entropy method to coarse-grain based on multiple reference atomistic simulations of short polypeptides with varied α and β character. Subsequently, we assess the CG model as a putative protein backbone forcefield by combining it with sidechain interactions based on native contacts but not incorporating native distances explicitly, unlike standard Go¯ models. We test the model's ability to fold a range of proteins and find that it achieves high accuracy (∼2 Å root mean square deviation resolution for both short sequences and large globular proteins), suggesting the strong role that backbone conformational preferences play in defining the fold landscape. This model can be systematically extended to non-natural amino acids and nonprotein polymers and sets the stage for extensions to non-Go¯ models with sequence-specific sidechain interactions.
Collapse
Affiliation(s)
- Tanmoy Sanyal
- Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | - Jeetain Mittal
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, USA
| | - M. Scott Shell
- Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, California 93106, USA
| |
Collapse
|
43
|
Andorfer R, Alper JD. From isolated structures to continuous networks: A categorization of cytoskeleton-based motile engineered biological microstructures. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1553. [PMID: 30740918 PMCID: PMC6881777 DOI: 10.1002/wnan.1553] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/27/2018] [Accepted: 12/28/2018] [Indexed: 11/06/2022]
Abstract
As technology at the small scale is advancing, motile engineered microstructures are becoming useful in drug delivery, biomedicine, and lab-on-a-chip devices. However, traditional engineering methods and materials can be inefficient or functionally inadequate for small-scale applications. Increasingly, researchers are turning to the biology of the cytoskeleton, including microtubules, actin filaments, kinesins, dyneins, myosins, and associated proteins, for both inspiration and solutions. They are engineering structures with components that range from being entirely biological to being entirely synthetic mimics of biology and on scales that range from isotropic continuous networks to single isolated structures. Motile biological microstructures trace their origins from the development of assays used to study the cytoskeleton to the array of structures currently available today. We define 12 types of motile biological microstructures, based on four categories: entirely biological, modular, hybrid, and synthetic, and three scales: networks, clusters, and isolated structures. We highlight some key examples, the unique functionalities, and the potential applications of each microstructure type, and we summarize the quantitative models that enable engineering them. By categorizing the diversity of motile biological microstructures in this way, we aim to establish a framework to classify these structures, define the gaps in current research, and spur ideas to fill those gaps. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Nanotechnology Approaches to Biology > Cells at the Nanoscale Biology-Inspired Nanomaterials > Protein and Virus-Based Structures Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Rachel Andorfer
- Department of Bioengineering, Clemson University, Clemson, South Carolina
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina
| | - Joshua D. Alper
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina
- Department of Biological Sciences, Clemson University, Clemson, South Carolina
- Eukaryotic Pathogen Innovations Center, Clemson University, Clemson, South Carolina
| |
Collapse
|
44
|
Consensus sequence design as a general strategy to create hyperstable, biologically active proteins. Proc Natl Acad Sci U S A 2019; 116:11275-11284. [PMID: 31110018 DOI: 10.1073/pnas.1816707116] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Consensus sequence design offers a promising strategy for designing proteins of high stability while retaining biological activity since it draws upon an evolutionary history in which residues important for both stability and function are likely to be conserved. Although there have been several reports of successful consensus design of individual targets, it is unclear from these anecdotal studies how often this approach succeeds and how often it fails. Here, we attempt to assess generality by designing consensus sequences for a set of six protein families with a range of chain lengths, structures, and activities. We characterize the resulting consensus proteins for stability, structure, and biological activities in an unbiased way. We find that all six consensus proteins adopt cooperatively folded structures in solution. Strikingly, four of six of these consensus proteins show increased thermodynamic stability over naturally occurring homologs. Each consensus protein tested for function maintained at least partial biological activity. Although peptide binding affinity by a consensus-designed SH3 is rather low, K m values for consensus enzymes are similar to values from extant homologs. Although consensus enzymes are slower than extant homologs at low temperature, they are faster than some thermophilic enzymes at high temperature. An analysis of sequence properties shows consensus proteins to be enriched in charged residues, and rarified in uncharged polar residues. Sequence differences between consensus and extant homologs are predominantly located at weakly conserved surface residues, highlighting the importance of these residues in the success of the consensus strategy.
Collapse
|
45
|
Leem J, Deane CM. High-Throughput Antibody Structure Modeling and Design Using ABodyBuilder. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2019; 1851:367-380. [PMID: 30298409 DOI: 10.1007/978-1-4939-8736-8_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Antibodies are proteins of the adaptive immune system; they can be designed to bind almost any molecule, and are increasingly being used as biotherapeutics. Experimental antibody design is an expensive and time-consuming process, and computational antibody design methods can now be used to help develop new therapeutics and diagnostics. Within the design pipeline, accurate antibody structure modeling is essential, as it provides the basis for antibody-antigen docking, binding affinity prediction, and estimating thermal stability. Ideally, models should be rapidly generated, allowing the exploration of the breadth of antibody space. This allows methods to replicate the natural processes of antibody diversification (e.g., V(D)J recombination and somatic hypermutation), and cope with large volumes of data that are typical of next-generation sequencing datasets. Here we describe ABodyBuilder and PEARS, algorithms that build and mutate antibody model structures. These methods take ~30 s to generate a model antibody structure.
Collapse
Affiliation(s)
- Jinwoo Leem
- Department of Statistics, University of Oxford, Oxford, UK
| | | |
Collapse
|
46
|
Abstract
The application of native enzymes may not be economical owing to the stability factor. A smaller protein molecule may be less susceptible to external stresses. Haloalkane dehalogenases (HLDs) that act on toxic haloalkanes may be incorporated as bioreceptors to detect haloalkane contaminants. Therefore, this study aims to develop mini proteins of HLD as an alternative bioreceptor which was able to withstand extreme conditions. Initially, the mini proteins were designed through computer modeling. Based on the results, five designed mini proteins were deemed to be viable stable mini proteins. They were then validated through experimental study. The smallest mini protein (model 5) was chosen for subsequent analysis as it was expressed in soluble form. No dehalogenase activity was detected, thus the specific binding interaction of between 1,3-dibromopropane with mini protein was investigated using isothermal titration calorimetry. Higher binding affinity between 1,3-dibromopropane and mini protein was obtained than the native. Thermal stability study with circular dichroism had proven that the mini protein possessed two times higher Tm value at 83.73 °C than the native at 43.97 °C. In conclusion, a stable mini protein was successfully designed and may be used as bioreceptors in the haloalkane sensor that is suitable for industrial application.
Collapse
|
47
|
García-Granados R, Lerma-Escalera JA, Morones-Ramírez JR. Metabolic Engineering and Synthetic Biology: Synergies, Future, and Challenges. Front Bioeng Biotechnol 2019; 7:36. [PMID: 30886847 PMCID: PMC6409320 DOI: 10.3389/fbioe.2019.00036] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/13/2019] [Indexed: 12/21/2022] Open
Abstract
The “-omics” era has brought a new set of tools and methods that have created a significant impact on the development of Metabolic Engineering and Synthetic Biology. These fields, rather than working separately, depend on each other to prosper and achieve their individual goals. Synthetic Biology aims to design libraries of genetic components (promoters, coding sequences, terminators, transcriptional factors and their binding sequences, and more), the assembly of devices, genetic circuits and even organism; in addition to obtaining quantitative information for the creation of models that can predict the behavior of biological systems (Cameron et al., 2014). Metabolic engineering seeks for the optimization of cellular processes, endemic to a specific organism, to produce a compound of interest from a substrate, preferably cheap and simple. It uses different databases, libraries of components and conditions to generate the maximum production rate of a desired chemical compound and avoiding inhibitors and conditions that affect the growth rate and other vital functions in the specific organism to achieve these goals; metabolic fluxes manipulation represents an important alternative (Stephanopoulos, 2012).
Collapse
Affiliation(s)
- Raúl García-Granados
- Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico.,Centro de Investigación en Biotecnología y Nanotecnología, Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, Apodaca, Mexico
| | - Jordy Alexis Lerma-Escalera
- Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico.,Centro de Investigación en Biotecnología y Nanotecnología, Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, Apodaca, Mexico.,Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| | - José R Morones-Ramírez
- Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico.,Centro de Investigación en Biotecnología y Nanotecnología, Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, Apodaca, Mexico
| |
Collapse
|
48
|
Simoncini D, Zhang KYJ, Schiex T, Barbe S. A structural homology approach for computational protein design with flexible backbone. Bioinformatics 2018; 35:2418-2426. [DOI: 10.1093/bioinformatics/bty975] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/01/2018] [Accepted: 11/28/2018] [Indexed: 01/09/2023] Open
Abstract
Abstract
Motivation
Structure-based Computational Protein design (CPD) plays a critical role in advancing the field of protein engineering. Using an all-atom energy function, CPD tries to identify amino acid sequences that fold into a target structure and ultimately perform a desired function. Energy functions remain however imperfect and injecting relevant information from known structures in the design process should lead to improved designs.
Results
We introduce Shades, a data-driven CPD method that exploits local structural environments in known protein structures together with energy to guide sequence design, while sampling side-chain and backbone conformations to accommodate mutations. Shades (Structural Homology Algorithm for protein DESign), is based on customized libraries of non-contiguous in-contact amino acid residue motifs. We have tested Shades on a public benchmark of 40 proteins selected from different protein families. When excluding homologous proteins, Shades achieved a protein sequence recovery of 30% and a protein sequence similarity of 46% on average, compared with the PFAM protein family of the target protein. When homologous structures were added, the wild-type sequence recovery rate achieved 93%.
Availability and implementation
Shades source code is available at https://bitbucket.org/satsumaimo/shades as a patch for Rosetta 3.8 with a curated protein structure database and ITEM library creation software.
Supplementary information
Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- David Simoncini
- Laboratoire d'Ingénierie des Systèmes Biologiques et des Procédés, LISBP, Université de Toulouse, CNRS, INRA, INSA, F Toulouse cedex 04, France
- Institut de recherche en informatique de Toulouse, IRIT, UMR 5505-CNRS, Université de Toulouse, Cedex 9, France
| | - Kam Y J Zhang
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, Yokohama, Kanagawa, Japan
| | - Thomas Schiex
- Institut de recherche en informatique de Toulouse, UMR 5505-CNRS, Université de Toulouse, Cedex 9, France
| | - Sophie Barbe
- Laboratoire d'Ingénierie des Systèmes Biologiques et des Procédés, LISBP, Université de Toulouse, CNRS, INRA, INSA, F Toulouse cedex 04, France
| |
Collapse
|
49
|
Song S, Ji J, Chen X, Gao S, Tang Z, Todo Y. Adoption of an improved PSO to explore a compound multi-objective energy function in protein structure prediction. Appl Soft Comput 2018. [DOI: 10.1016/j.asoc.2018.07.042] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
50
|
Wong SWK, Liu JS, Kou SC. Exploring the conformational space for protein folding with sequential Monte Carlo. Ann Appl Stat 2018. [DOI: 10.1214/17-aoas1124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|