1
|
Sinha SK, Gholap A, C M Y, Pal A, Kapdi AR, Maiti D. Generating the Pd-catalyzed δ C-H chalcogenation of aliphatic picolinamides: systematically decreasing the bias. Chem Sci 2025; 16:7936-7945. [PMID: 40191127 PMCID: PMC11969726 DOI: 10.1039/d4sc07897g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/16/2025] [Indexed: 04/09/2025] Open
Abstract
Reaching out to the distal sp3 C-H bonds remains a daunting challenge to synthetic organic chemists, primarily due to the relative inertness of the C-H bonds in alkanes. As such, most reports have envisaged the employment of sterically biased substrates, which render the other possible positions inaccessible for functionalization. Herein, we report a palladium-catalyzed highly selective δ-chalcogenation of aliphatic picolinamides, whereby both sterically biased and relatively unbiased substrates are made feasible for site-selective δ-C-H functionalization. The successful employment of the Thorpe-Ingold effect explains the reactive intermediates involved. The present protocol also provides direct access to the introduction of structural modifications on α-amino acid structural motifs, such as leucine, with high regioselectivity. Sequential hetero-di-functionalization has been carried out at δ-sp3 C-H bonds, resulting in the desymmetrization of quaternary centers. A thorough mechanistic investigation has been carried out, which provided evidence for the reaction pathway and the plausible mechanistic cycle involved.
Collapse
Affiliation(s)
- Soumya Kumar Sinha
- Department of Chemistry, Indian Institute of Technology Bombay Powai Mumbai 400076 India
| | - Aniket Gholap
- Department of Chemistry, Institute of Chemical Technology Nathalal Parekh Road, Matunga Mumbai-400019 India
| | - Yazhinimuthu C M
- Department of Chemistry, Indian Institute of Technology Bombay Powai Mumbai 400076 India
| | - Anirban Pal
- Department of Chemistry, Indian Institute of Technology Bombay Powai Mumbai 400076 India
| | - Anant R Kapdi
- Department of Chemistry, Institute of Chemical Technology Nathalal Parekh Road, Matunga Mumbai-400019 India
| | - Debabrata Maiti
- Department of Chemistry, Indian Institute of Technology Bombay Powai Mumbai 400076 India
| |
Collapse
|
2
|
Møller CHS, Sauer SPA. RPA(D) and HRPA(D): Calculating NMR Spin-Spin Coupling Constants in Free Amino Acid Residues. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2025; 63:328-345. [PMID: 39950736 DOI: 10.1002/mrc.5514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/15/2025] [Accepted: 01/23/2025] [Indexed: 03/05/2025]
Abstract
In the pursuit of computational methods which boast both low computational cost and a high degree of accuracy, the SOPPA-derived methods RPA(D) and HRPA(D) are showing great promise. This study aims to further the benchmarking of these two methods in comparison with both the original SOPPA and the CCSD method by calculating NMR spin-spin coupling constants in the backbone structure of free amino acid residues. Based on a small basis set study, the relative performance of the methods was not found to be heavily dependent on the size of the basis set. While HRPA(D) was found to reproduce the SOPPA results to a consistently high degree of accuracy, RPA(D) reproduced the CCSD results for the one-bond coupling constants more accurately than both HRPA(D) and SOPPA.
Collapse
Affiliation(s)
| | - Stephan P A Sauer
- Department of Chemistry, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Pilcher C, Buco PAV, Truong JQ, Ramsland PA, Smeets MF, Walkley CR, Holien JK. Characteristics of the Kelch domain containing (KLHDC) subfamily and relationships with diseases. FEBS Lett 2025; 599:1094-1112. [PMID: 39887712 PMCID: PMC12035522 DOI: 10.1002/1873-3468.15108] [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: 10/28/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 02/01/2025]
Abstract
The Kelch protein superfamily is an evolutionary conserved family containing 63 alternate protein coding members. The superfamily is split into three subfamilies: Kelch like (KLHL), Kelch-repeat and bric-a-bracs (BTB) domain containing (KBTBD) and Kelch domain containing protein (KLHDC). The KLHDC subfamily is one of the smallest within the Kelch superfamily, containing 10 primary members. There is little known about the structures and functions of the subfamily; however, they are thought to be involved in several cellular and molecular processes. Recently, there have been significant structural and biochemical advances for KLHDC2, which has aided our understanding of other KLHDC family members. Furthermore, small molecules directly targeting KLHDC2 have been identified, which act as tools for targeted protein degradation. This review utilises this information, in conjunction with a thorough exploration of the structural aspects and potential biological functions to summarise the relationship between KLHDCs and human disease.
Collapse
Affiliation(s)
- Courtney Pilcher
- School of Science, STEM CollegeRMIT UniversityMelbourneAustralia
- St Vincent's Institute of Medical ResearchFitzroyAustralia
| | - Paula Armina V. Buco
- St Vincent's Institute of Medical ResearchFitzroyAustralia
- Department of Medicine, Eastern Hill Academic Centre, Melbourne Medical SchoolThe University of MelbourneCarltonAustralia
| | - Jia Q. Truong
- School of Science, STEM CollegeRMIT UniversityMelbourneAustralia
| | - Paul A. Ramsland
- School of Science, STEM CollegeRMIT UniversityMelbourneAustralia
- Department of ImmunologyMonash UniversityMelbourneAustralia
- Department of Surgery, Austin HealthThe University of MelbourneMelbourneAustralia
| | | | - Carl R. Walkley
- St Vincent's Institute of Medical ResearchFitzroyAustralia
- Department of Medicine, Eastern Hill Academic Centre, Melbourne Medical SchoolThe University of MelbourneCarltonAustralia
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchMelbourneAustralia
- Department of Molecular and Translational ScienceMonash UniversityMelbourneAustralia
| | - Jessica K. Holien
- School of Science, STEM CollegeRMIT UniversityMelbourneAustralia
- St Vincent's Institute of Medical ResearchFitzroyAustralia
- Department of Medicine, Eastern Hill Academic Centre, Melbourne Medical SchoolThe University of MelbourneCarltonAustralia
| |
Collapse
|
4
|
Liu X, Hu B, Yu Z. Noncanonical Amino Acids Dictate Peptide Assembly in Living Cells. Acc Chem Res 2025; 58:1081-1093. [PMID: 40105513 DOI: 10.1021/acs.accounts.4c00796] [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: 03/20/2025]
Abstract
ConspectusEmulating the structural features or functions of natural systems has been demonstrated as a state-of-the-art strategy to create artificial functional materials. Inspired by the assembly and bioactivity of proteins, the self-assembly of peptides into nanostructures represents a promising approach for creating biomaterials. Conventional assembled peptide biomaterials are typically formulated in solution and delivered to pathological sites for implementing theranostic objectives. However, this translocation entails a switch from formulation conditions to the physiological environment and raises concerns about material performance. In addition, the precise and efficient accumulation of administered biomaterials at target sites remains a significant challenge, leading to potential biosafety issues associated with off-target effects. These limitations significantly hinder the progress of advanced biomaterials. To address these concerns, the past few years have witnessed the development of in situ assembly of peptides in living systems as a new endeavor for optimizing biomaterial performance benefiting from the advances of stimuli-responsive reactions regulating noncovalent interactions. In situ assembly of peptides refers to the processes of regulating assembly via stimuli-responsive reactions at target sites. Due to the advantages of precisely forming well-defined nanostructures at pathological lesions, in situ-formed assemblies with integrated bioactivity are interesting for the development of next-generation biomedical agents.Despite the great potential of in situ assembly of peptides for developing biomedical agents, this research area still suffers from a limited toolkit for operating peptide assembly under complicated physiological conditions. Considering the advantages of amino acids in being incorporated into peptide backbones and modified with stimuli-responsive units, development of an amino acid toolkit is promising to address this concern. Therefore, our laboratory has been intensively engaged in designing and discovering stimuli-responsive noncanonical amino acids (ncAAs) to expand the toolkit for manipulating peptide assembly under various biological conditions. Thus far, we have synthesized peptides containing ncAAs 4-aminoproline, 2-nitroimidazole alanine, Se-methionine, sulfated tyrosine, and glycosylated serine, which allow us to develop acid-responsive, redox-responsive, and enzyme-responsive assembly systems. Based on these stimuli-responsive ncAAs, we have established complex self-sorting assembly, self-amplified assembly, and dissipative assembly systems in living cells to optimize the bioactivity of peptides. The resulting in situ assembly systems exhibit morphological adaptability to the biological microenvironment, which contributes to overcoming delivery barriers and improvement of targeting accumulation. Therefore, by utilizing the developed toolkit, we have further created supramolecular PROTACs, supramolecular antagonists, and supramolecular probes for cancer treatment and diagnosis to highlight the implications of ncAAs for biomedical usage. In this Account, we summarize our journey of in situ self-assembly of peptides in living cells utilizing stimuli-responsive ncAAs, with an emphasis on the mechanism for regulating peptide assembly and optimizing the bioactivity of peptides. Eventually, we also provide our forward conceiving prospects on the challenges for the further development of in situ assembly of peptides in living systems and the clinical translation of in situ-formulated biomaterials.
Collapse
Affiliation(s)
- Xin Liu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Binbin Hu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Zhilin Yu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
5
|
Chen Y, Clay N, Phan N, Lothrop E, Culkins C, Robinson B, Stubblefield A, Ferguson A, Kimmel BR. Molecular Matchmakers: Bioconjugation Techniques Enhance Prodrug Potency for Immunotherapy. Mol Pharm 2025; 22:58-80. [PMID: 39570179 DOI: 10.1021/acs.molpharmaceut.4c00867] [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: 11/22/2024]
Abstract
Cancer patients suffer greatly from the severe off-target side effects of small molecule drugs, chemotherapy, and radiotherapy─therapies that offer little protection following remission. Engineered immunotherapies─including cytokines, immune checkpoint blockade, monoclonal antibodies, and CAR-T cells─provide better targeting and future tumor growth prevention. Still, issues such as ineffective activation, immunogenicity, and off-target effects remain primary concerns. "Prodrug" therapies─classified as therapies administered as inactive and then selectively activated to control the time and area of release─hold significant promise in overcoming these concerns. Bioconjugation techniques (e.g., natural linker conjugation, bioorthogonal reactions, and noncanonical amino acid incorporation) enable the rapid and homogeneous synthesis of prodrugs and offer selective loading of immunotherapeutic agents to carrier molecules and protecting groups to prevent off-target effects after administration. Several prodrug activation mechanisms have been highlighted for cancer therapeutics, including endogenous activation by hypoxic or acidic conditions common in tumors, exogenous activation by targeted bioorthogonal cleavage, or stimuli-responsive light activation, and dual-stimuli activation, which adds specificity by combining these mechanisms. This review will explore modern prodrug conjugation and activation options, focusing on how these strategies can enhance immunotherapy responses and improve patient outcomes. We will also discuss the implications of computational methodology for therapy design and recommend procedures to determine how and where to conjugate carrier systems and "prodrug" groups onto therapeutic agents to enhance the safety and control of these delivery platforms.
Collapse
Affiliation(s)
- Yinuo Chen
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Natalie Clay
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Nathan Phan
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Elijah Lothrop
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Courtney Culkins
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise Robinson
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ariana Stubblefield
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Alani Ferguson
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise R Kimmel
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Engineering, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
6
|
Puppala A, Sosa D, Castillo Suchkou J, French R, Dobosz-Bartoszek M, Kiernan K, Simonović M. Human selenocysteine synthase, SEPSECS, has evolved to optimize binding of a tRNA-based substrate. Nucleic Acids Res 2024; 52:13368-13385. [PMID: 39385655 PMCID: PMC11602143 DOI: 10.1093/nar/gkae875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024] Open
Abstract
The evolution of the genetic code to incorporate selenocysteine (Sec) enabled the development of a selenoproteome in all domains of life. O-phosphoseryl-tRNASec selenium transferase (SepSecS) catalyzes the terminal reaction of Sec synthesis on tRNASec in archaea and eukaryotes. Despite harboring four equivalent active sites, human SEPSECS binds no more than two tRNASec molecules. Though, the basis for this asymmetry remains poorly understood. In humans, an acidic, C-terminal, α-helical extension precludes additional tRNA-binding events in two of the enzyme monomers, stabilizing the SEPSECS•tRNASec complex. However, the existence of a helix exclusively in vertebrates raised questions about the evolution of the tRNA-binding mechanism in SEPSECS and the origin of its C-terminal extension. Herein, using a comparative structural and phylogenetic analysis, we show that the tRNA-binding motifs in SEPSECS are poorly conserved across species. Consequently, in contrast to mammalian SEPSECS, the archaeal ortholog cannot bind unacylated tRNASec and requires an aminoacyl group. Moreover, the C-terminal α-helix 16 is a mammalian innovation, and its absence causes aggregation of the SEPSECS•tRNASec complex at low tRNA concentrations. Altogether, we propose SEPSECS evolved a tRNASec binding mechanism as a crucial functional and structural feature, allowing for additional levels of regulation of Sec and selenoprotein synthesis.
Collapse
Affiliation(s)
- Anupama K Puppala
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Dylan Sosa
- Department of Ecology & Evolution, University of Chicago, Chicago, IL 60637, USA
| | - Jennifer Castillo Suchkou
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Rachel L French
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Malgorzata Dobosz-Bartoszek
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Kaitlyn A Kiernan
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Miljan Simonović
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
7
|
Lino BR, Williams SJ, Castor ME, Van Deventer JA. Reaching New Heights in Genetic Code Manipulation with High Throughput Screening. Chem Rev 2024; 124:12145-12175. [PMID: 39418482 PMCID: PMC11879460 DOI: 10.1021/acs.chemrev.4c00329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The chemical and physical properties of proteins are limited by the 20 canonical amino acids. Genetic code manipulation allows for the incorporation of noncanonical amino acids (ncAAs) that enhance or alter protein functionality. This review explores advances in the three main strategies for introducing ncAAs into biosynthesized proteins, focusing on the role of high throughput screening in these advancements. The first section discusses engineering aminoacyl-tRNA synthetases (aaRSs) and tRNAs, emphasizing how novel selection methods improve characteristics including ncAA incorporation efficiency and selectivity. The second section examines high-throughput techniques for improving protein translation machinery, enabling accommodation of alternative genetic codes. This includes opportunities to enhance ncAA incorporation through engineering cellular components unrelated to translation. The final section highlights various discovery platforms for high-throughput screening of ncAA-containing proteins, showcasing innovative binding ligands and enzymes that are challenging to create with only canonical amino acids. These advances have led to promising drug leads and biocatalysts. Overall, the ability to discover unexpected functionalities through high-throughput methods significantly influences ncAA incorporation and its applications. Future innovations in experimental techniques, along with advancements in computational protein design and machine learning, are poised to further elevate this field.
Collapse
Affiliation(s)
- Briana R. Lino
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| | - Sean J. Williams
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| | - Michelle E. Castor
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| | - James A. Van Deventer
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
- Biomedical Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
8
|
Bhattacharjee R, Lemke EA. Potential vs Challenges of Expanding the Protein Universe With Genetic Code Expansion in Eukaryotic Cells. J Mol Biol 2024; 436:168807. [PMID: 39357814 DOI: 10.1016/j.jmb.2024.168807] [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] [Received: 07/14/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Following decades of innovation and perfecting, genetic code expansion has become a powerful tool for in vivo protein modification. Some of the major hurdles that had to be overcome include suboptimal performance of GCE-specific translational components in host systems, competing cellular processes, unspecific modification of the host proteome and limited availability of codons for reassignment. Although strategies have been developed to overcome these challenges, there is critical need for further advances. Here we discuss the current state-of-the-art in genetic code expansion technology and the issues that still need to be addressed to unleash the full potential of this method in eukaryotic cells.
Collapse
Affiliation(s)
- Rajanya Bhattacharjee
- Biocenter, Johannes Gutenberg University Mainz, 55128, Mainz, Germany; IMB International PhD Programme (IPP), 55128 Mainz, Germany
| | - Edward A Lemke
- Biocenter, Johannes Gutenberg University Mainz, 55128, Mainz, Germany; Institute of Molecular Biology (IMB), 55128 Mainz, Germany.
| |
Collapse
|
9
|
Yang S, Jin S, Zhang M, Chen Y, Guo Y, Hu Y, Wolynes PG, Xiao H. Real-Time Visualization of Protein Microenvironment Changes with High Spatial Resolution in Live Cells via Site-Specific Incorporation of Rotor-Based Fluorescent Noncanonical Amino Acids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.19.619218. [PMID: 39484402 PMCID: PMC11526926 DOI: 10.1101/2024.10.19.619218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Traditional methods, such as the use of fluorescent protein fusions and environment-sensitive fluorophores, have limitations when studying protein microenvironment changes at the finest spatial resolution. These techniques often rely on bulky proteins or tags restricted to the N- or C-terminus, which can disrupt the natural behavior of the target protein and dramatically limit the ability of their method to investigate noninvasively microenvironment effects. To overcome these challenges, we have developed an innovative strategy to visualize microenvironment changes of protein substructures in real-time by genetically incorporating environment-sensitive noncanonical amino acids (ncAAs) containing rotor-based fluorophores (RBFs) at specific positions within a protein of interest. Through computational redesign of aminoacyl-tRNA synthetase, we successfully incorporated these rotor-based ncAAs into various proteins in mammalian cells. By site-specifically placing these ncAAs in distinct regions of proteins, we detected microenvironmental changes of several different protein domains during events such as aggregation, clustering, aggregation disassembly, and cluster dissociation.
Collapse
|
10
|
Guo Y, Cheng L, Hu Y, Zhang M, Liu R, Wang Y, Jiang S, Xiao H. Biosynthesis of Halogenated Tryptophans for Protein Engineering Using Genetic Code Expansion. Chembiochem 2024; 25:e202400366. [PMID: 38958600 PMCID: PMC11483216 DOI: 10.1002/cbic.202400366] [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: 04/20/2024] [Revised: 06/29/2024] [Accepted: 07/03/2024] [Indexed: 07/04/2024]
Abstract
Genetic Code Expansion technology offers significant potential in incorporating noncanonical amino acids into proteins at precise locations, allowing for the modulation of protein structures and functions. However, this technology is often limited by the need for costly and challenging-to-synthesize external noncanonical amino acid sources. In this study, we address this limitation by developing autonomous cells capable of biosynthesizing halogenated tryptophan derivatives and introducing them into proteins using Genetic Code Expansion technology. By utilizing inexpensive halide salts and different halogenases, we successfully achieve the selective biosynthesis of 6-chloro-tryptophan, 7-chloro-tryptophan, 6-bromo-tryptophan, and 7-bromo-tryptophan. These derivatives are introduced at specific positions with corresponding bioorthogonal aminoacyl-tRNA synthetase/tRNA pairs in response to the amber codon. Following optimization, we demonstrate the robust expression of proteins containing halogenated tryptophan residues in cells with the ability to biosynthesize these tryptophan derivatives. This study establishes a versatile platform for engineering proteins with various halogenated tryptophans.
Collapse
Affiliation(s)
- Yiming Guo
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005, U.S.A
| | - Linqi Cheng
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005, U.S.A
| | - Yu Hu
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005, U.S.A
| | - Mengxi Zhang
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005, U.S.A
| | - Rui Liu
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005, U.S.A
| | - Yixian Wang
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005, U.S.A
| | - Shiyu Jiang
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005, U.S.A
| | - Han Xiao
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005, U.S.A
- SynthX Center, Rice University, 6100 Main Street, Houston, Texas, 77005, U.S.A
- Department of Biosciences, Rice University, 6100 Main Street, Houston, Texas, 77005, U.S.A
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005, USA
| |
Collapse
|
11
|
Tennakoon R, Cui H. Aminoacyl-tRNA synthetases. Curr Biol 2024; 34:R884-R888. [PMID: 39378843 DOI: 10.1016/j.cub.2024.08.029] [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: 10/10/2024]
Abstract
Aminoacyl-tRNA synthetases hold the key to the genetic code and assign nucleic acid-based codons to amino acids, the building blocks of proteins. In their ability to recognize identity elements on transfer RNAs (tRNAs), some as simple as a single base pair, they ensure that the same proteins are formed each time information embedded in DNA is transcribed into messenger RNA (mRNA) and translated into proteins (Figure 1A). Thus, aminoacyl-tRNA synthetase active sites are conserved; however, since their evolutionary origin, their functions have been co-opted, expanded on and played novel roles during evolution. Below, we provide an overview of the many functions of aminoacyl-tRNA synthetases - from their role in translation, one of the most fundamental processes of all life, to newly discovered, diverse functions.
Collapse
Affiliation(s)
| | - Haissi Cui
- Department of Chemistry, University of Toronto, Toronto, Canada.
| |
Collapse
|
12
|
Jin WY, Guo JX, Tang R, Wang J, Zhao H, Zhang M, Teng LZ, Sansonetti PJ, Gao YZ. In vivo detection of endogenous toxic phenolic compounds of intestine. JOURNAL OF HAZARDOUS MATERIALS 2024; 478:135526. [PMID: 39153300 DOI: 10.1016/j.jhazmat.2024.135526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Phenol and p-cresol are two common toxic small molecules related to various diseases. Existing reports confirmed that high L-tyrosine in the daily diet can increase the concentration of phenolic compounds in blood and urine. L-tyrosine is a common component of protein-rich foods. Some anaerobic bacteria in the gut can convert non-toxic l-tyrosine into these two toxic phenolic compounds, phenol and p-cresol. Existing methods have been constructed for measuring the concentration of phenolic compound in feces. However, there is still a lack of direct visual evidence to measure the phenolic compounds in the intestine. In this study, we aimed to construct a whole-cell biosensor for phenolic compounds detection based on the dmpR, the regulator from the phenol metabolism cluster. The commensal bacterium Citrobacter amalonaticus PS01 was selected and used as the chassis. Compared with the biosensor based on ECN1917, the biosensor PS01[dmpR] could better implant into the mouse gut through gavage and showed a higher sensitive to phenolic compound. And the concentration of phenolic compounds in the intestines could be observed with the help of in vivo imaging system using PS01[dmpR]. This paper demonstrated endogenous phenol synthesis in the gut and the strategy of using commensal bacteria to construct whole-cell biosensors for detecting small molecule compounds in the intestines.
Collapse
Affiliation(s)
- Wen-Yu Jin
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jia-Xin Guo
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Rongkang Tang
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jielin Wang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huan Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meng Zhang
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; Pasteurian College, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Lin-Zuo Teng
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Philippe J Sansonetti
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Université de Paris, Paris, France.
| | - Yi-Zhou Gao
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
13
|
Jann C, Giofré S, Bhattacharjee R, Lemke EA. Cracking the Code: Reprogramming the Genetic Script in Prokaryotes and Eukaryotes to Harness the Power of Noncanonical Amino Acids. Chem Rev 2024; 124:10281-10362. [PMID: 39120726 PMCID: PMC11441406 DOI: 10.1021/acs.chemrev.3c00878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 06/10/2024] [Accepted: 06/27/2024] [Indexed: 08/10/2024]
Abstract
Over 500 natural and synthetic amino acids have been genetically encoded in the last two decades. Incorporating these noncanonical amino acids into proteins enables many powerful applications, ranging from basic research to biotechnology, materials science, and medicine. However, major challenges remain to unleash the full potential of genetic code expansion across disciplines. Here, we provide an overview of diverse genetic code expansion methodologies and systems and their final applications in prokaryotes and eukaryotes, represented by Escherichia coli and mammalian cells as the main workhorse model systems. We highlight the power of how new technologies can be first established in simple and then transferred to more complex systems. For example, whole-genome engineering provides an excellent platform in bacteria for enabling transcript-specific genetic code expansion without off-targets in the transcriptome. In contrast, the complexity of a eukaryotic cell poses challenges that require entirely new approaches, such as striving toward establishing novel base pairs or generating orthogonally translating organelles within living cells. We connect the milestones in expanding the genetic code of living cells for encoding novel chemical functionalities to the most recent scientific discoveries, from optimizing the physicochemical properties of noncanonical amino acids to the technological advancements for their in vivo incorporation. This journey offers a glimpse into the promising developments in the years to come.
Collapse
Affiliation(s)
- Cosimo Jann
- Biocenter, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
- IMB
Postdoc Programme (IPPro), 55128 Mainz, Germany
| | - Sabrina Giofré
- Biocenter, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
- IMB
Postdoc Programme (IPPro), 55128 Mainz, Germany
| | - Rajanya Bhattacharjee
- Biocenter, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
- IMB
International PhD Programme (IPP), 55128 Mainz, Germany
| | - Edward A. Lemke
- Biocenter, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
- Institute
of Molecular Biology (IMB), 55128 Mainz, Germany
| |
Collapse
|
14
|
Söllner J, Derler I. Genetic code expansion, an emerging tool in the Ca 2+ ion channel field. J Physiol 2024; 602:3297-3313. [PMID: 38695316 DOI: 10.1113/jp285840] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/15/2024] [Indexed: 07/17/2024] Open
Abstract
Various methods for characterizing binding forces as well as for monitoring and remote control of ion channels are still emerging. A recent innovation is the direct incorporation of unnatural amino acids (UAAs) with corresponding biophysical or biochemical properties, which are integrated using genetic code expansion technology. Minimal changes to natural amino acids, which are achieved by chemical synthesis of corresponding UAAs, are valuable tools to provide insight into the contributions of physicochemical properties of side chains in binding events. To gain unique control over the conformational changes or function of ion channels, a series of light-sensitive, chemically reactive and posttranslationally modified UAAs have been developed and utilized. Here, we present the existing UAA tools, their mode of action, their potential and limitations as well as their previous applications to Ca2+-permeable ion channels.
Collapse
Affiliation(s)
- Julia Söllner
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz, Austria
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz, Austria
| |
Collapse
|
15
|
Evans R, Krahn N, Weiss J, Vincent KA, Söll D, Armstrong FA. Replacing a Cysteine Ligand by Selenocysteine in a [NiFe]-Hydrogenase Unlocks Hydrogen Production Activity and Addresses the Role of Concerted Proton-Coupled Electron Transfer in Electrocatalytic Reversibility. J Am Chem Soc 2024; 146:16971-16976. [PMID: 38747098 PMCID: PMC11212049 DOI: 10.1021/jacs.4c03489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 06/27/2024]
Abstract
Hydrogenases catalyze hydrogen/proton interconversion that is normally electrochemically reversible (having minimal overpotential requirement), a special property otherwise almost exclusive to platinum metals. The mechanism of [NiFe]-hydrogenases includes a long-range proton-coupled electron-transfer process involving a specific Ni-coordinated cysteine and the carboxylate of a nearby glutamate. A variant in which this cysteine has been exchanged for selenocysteine displays two distinct changes in electrocatalytic properties, as determined by protein film voltammetry. First, proton reduction, even in the presence of H2 (a strong product inhibitor), is greatly enhanced relative to H2 oxidation: this result parallels a characteristic of natural [NiFeSe]-hydrogenases which are superior H2 production catalysts. Second, an inflection (an S-shaped "twist" in the trace) appears around the formal potential, the small overpotentials introduced in each direction (oxidation and reduction) signaling a departure from electrocatalytic reversibility. Concerted proton-electron transfer offers a lower energy pathway compared to stepwise transfers. Given the much lower proton affinity of Se compared to that of S, the inflection provides compelling evidence that concerted proton-electron transfer is important in determining why [NiFe]-hydrogenases are reversible electrocatalysts.
Collapse
Affiliation(s)
- Rhiannon
M. Evans
- Department
of Chemistry, University of Oxford, Oxford OX1 3TA, United Kingdom
| | - Natalie Krahn
- Department
of Biochemistry and Molecular Biology, University
of Georgia, Athens, Georgia 30602, United
States
| | - Joshua Weiss
- Department
of Biochemistry and Molecular Biology, University
of Georgia, Athens, Georgia 30602, United
States
| | - Kylie A. Vincent
- Department
of Chemistry, University of Oxford, Oxford OX1 3TA, United Kingdom
| | - Dieter Söll
- Department
of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06511, United States
- Department
of Chemistry, Yale University, New Haven, Connecticut 06520, United States
| | - Fraser A. Armstrong
- Department
of Chemistry, University of Oxford, Oxford OX1 3TA, United Kingdom
| |
Collapse
|
16
|
Barat A, Powner MW. Spontaneous Peptide Ligation Mediated by Cysteamine. JACS AU 2024; 4:1752-1757. [PMID: 38818061 PMCID: PMC11134366 DOI: 10.1021/jacsau.4c00243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 06/01/2024]
Abstract
The fundamental and universal nature of life's exploitation of peptides suggests they must have played a vital role during the onset of life, but their spontaneous chemoselective synthesis in water remains unknown. Aminonitriles (1) are widely accepted as prebiotic precursors of both amino acids and peptides, but they do not spontaneously polymerize in water to yield peptides. Here, we demonstrate that the simple prebiotically plausible aminothiol, cysteamine (5), participates in Strecker chemistry to furnish β-mercaptoethyl-α-aminonitriles (8) and β-mercaptoethyl-amino acids (16), which are predisposed to spontaneously form peptides in water. Intramolecular thiol catalyzed ligation is faster, higher-yielding, and more α-selective than previously reported prebiotic peptide ligation chemistries, enabling, for example, the highly regioselective α-ligation of Asp- and Glu-dinitriles in quantitative yields. Our findings suggest that cysteamine (5), the thiol bearing moiety of the universal thiol cofactor coenzyme A, may have played an important role in the selective chemical synthesis of prebiotic α-peptides.
Collapse
Affiliation(s)
- Abid Barat
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, United Kingdom
| | - Matthew W. Powner
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, United Kingdom
| |
Collapse
|
17
|
Sigal M, Matsumoto S, Beattie A, Katoh T, Suga H. Engineering tRNAs for the Ribosomal Translation of Non-proteinogenic Monomers. Chem Rev 2024; 124:6444-6500. [PMID: 38688034 PMCID: PMC11122139 DOI: 10.1021/acs.chemrev.3c00894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/21/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024]
Abstract
Ribosome-dependent protein biosynthesis is an essential cellular process mediated by transfer RNAs (tRNAs). Generally, ribosomally synthesized proteins are limited to the 22 proteinogenic amino acids (pAAs: 20 l-α-amino acids present in the standard genetic code, selenocysteine, and pyrrolysine). However, engineering tRNAs for the ribosomal incorporation of non-proteinogenic monomers (npMs) as building blocks has led to the creation of unique polypeptides with broad applications in cellular biology, material science, spectroscopy, and pharmaceuticals. Ribosomal polymerization of these engineered polypeptides presents a variety of challenges for biochemists, as translation efficiency and fidelity is often insufficient when employing npMs. In this Review, we will focus on the methodologies for engineering tRNAs to overcome these issues and explore recent advances both in vitro and in vivo. These efforts include increasing orthogonality, recruiting essential translation factors, and creation of expanded genetic codes. After our review on the biochemical optimizations of tRNAs, we provide examples of their use in genetic code manipulation, with a focus on the in vitro discovery of bioactive macrocyclic peptides containing npMs. Finally, an analysis of the current state of tRNA engineering is presented, along with existing challenges and future perspectives for the field.
Collapse
Affiliation(s)
- Maxwell Sigal
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Satomi Matsumoto
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Adam Beattie
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takayuki Katoh
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroaki Suga
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
18
|
Rozhoňová H, Martí-Gómez C, McCandlish DM, Payne JL. Robust genetic codes enhance protein evolvability. PLoS Biol 2024; 22:e3002594. [PMID: 38754362 PMCID: PMC11098591 DOI: 10.1371/journal.pbio.3002594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 03/19/2024] [Indexed: 05/18/2024] Open
Abstract
The standard genetic code defines the rules of translation for nearly every life form on Earth. It also determines the amino acid changes accessible via single-nucleotide mutations, thus influencing protein evolvability-the ability of mutation to bring forth adaptive variation in protein function. One of the most striking features of the standard genetic code is its robustness to mutation, yet it remains an open question whether such robustness facilitates or frustrates protein evolvability. To answer this question, we use data from massively parallel sequence-to-function assays to construct and analyze 6 empirical adaptive landscapes under hundreds of thousands of rewired genetic codes, including those of codon compression schemes relevant to protein engineering and synthetic biology. We find that robust genetic codes tend to enhance protein evolvability by rendering smooth adaptive landscapes with few peaks, which are readily accessible from throughout sequence space. However, the standard genetic code is rarely exceptional in this regard, because many alternative codes render smoother landscapes than the standard code. By constructing low-dimensional visualizations of these landscapes, which each comprise more than 16 million mRNA sequences, we show that such alternative codes radically alter the topological features of the network of high-fitness genotypes. Whereas the genetic codes that optimize evolvability depend to some extent on the detailed relationship between amino acid sequence and protein function, we also uncover general design principles for engineering nonstandard genetic codes for enhanced and diminished evolvability, which may facilitate directed protein evolution experiments and the bio-containment of synthetic organisms, respectively.
Collapse
Affiliation(s)
- Hana Rozhoňová
- Institute of Integrative Biology, ETH Zürich, Zürich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Carlos Martí-Gómez
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - David M. McCandlish
- Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Joshua L. Payne
- Institute of Integrative Biology, ETH Zürich, Zürich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
19
|
Yuan Y, Li P, Li J, Zhao Q, Chang Y, He X. Protein lipidation in health and disease: molecular basis, physiological function and pathological implication. Signal Transduct Target Ther 2024; 9:60. [PMID: 38485938 PMCID: PMC10940682 DOI: 10.1038/s41392-024-01759-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/31/2023] [Accepted: 01/24/2024] [Indexed: 03/18/2024] Open
Abstract
Posttranslational modifications increase the complexity and functional diversity of proteins in response to complex external stimuli and internal changes. Among these, protein lipidations which refer to lipid attachment to proteins are prominent, which primarily encompassing five types including S-palmitoylation, N-myristoylation, S-prenylation, glycosylphosphatidylinositol (GPI) anchor and cholesterylation. Lipid attachment to proteins plays an essential role in the regulation of protein trafficking, localisation, stability, conformation, interactions and signal transduction by enhancing hydrophobicity. Accumulating evidence from genetic, structural, and biomedical studies has consistently shown that protein lipidation is pivotal in the regulation of broad physiological functions and is inextricably linked to a variety of diseases. Decades of dedicated research have driven the development of a wide range of drugs targeting protein lipidation, and several agents have been developed and tested in preclinical and clinical studies, some of which, such as asciminib and lonafarnib are FDA-approved for therapeutic use, indicating that targeting protein lipidations represents a promising therapeutic strategy. Here, we comprehensively review the known regulatory enzymes and catalytic mechanisms of various protein lipidation types, outline the impact of protein lipidations on physiology and disease, and highlight potential therapeutic targets and clinical research progress, aiming to provide a comprehensive reference for future protein lipidation research.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peiyuan Li
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianghui Li
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China.
| | - Ying Chang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China.
| | - Xingxing He
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China.
| |
Collapse
|
20
|
Tang YD, Yu C, Cai XH. Novel technologies are turning a dream into reality: conditionally replicating viruses as vaccines. Trends Microbiol 2024; 32:292-301. [PMID: 37798168 DOI: 10.1016/j.tim.2023.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 10/07/2023]
Abstract
Conditionally replicating viruses (CRVs) are a type of virus with one or more essential gene functions that are impaired resulting in the disruption of viral genome replication, protein synthesis, or virus particle assembly. CRVs can replicate only if the deficient essential genes are supplied. CRVs are widely used in biomedical research, particularly as vaccines. Traditionally, CRVs are generated by creating complementary cell lines that provide the impaired genes. With the development of biotechnology, novel techniques have been invented to generate CRVs, such as targeted protein degradation (TPD) technologies and premature termination codon (PTC) read-through technologies. The advantages and disadvantages of these novel technologies are discussed. Finally, we provide perspectives on what challenges need to be overcome for CRVs to reach the market.
Collapse
Affiliation(s)
- Yan-Dong Tang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China; Heilongjiang Provincial Research Center for Veterinary Biomedicine, Harbin, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, China.
| | - Changqing Yu
- Engineering Center of Agricultural Biosafety Assessment and Biotechnology, School of Advanced Agricultural Sciences, Yibin Vocational and Technical College, Yibin, China.
| | - Xue-Hui Cai
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China; Heilongjiang Provincial Research Center for Veterinary Biomedicine, Harbin, China.
| |
Collapse
|
21
|
Debnath T, Cisneros GA. Investigation of the stability of D5SIC-DNAM-incorporated DNA duplex in Taq polymerase binary system: a systematic classical MD approach. Phys Chem Chem Phys 2024; 26:7287-7295. [PMID: 38353000 PMCID: PMC11078294 DOI: 10.1039/d3cp05571j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
DNA polymerases are fundamental enzymes that play a crucial role in processing DNA with high fidelity and accuracy ensuring the faithful transmission of genetic information. The recognition of unnatural base pairs (UBPs) by polymerases, enabling their replication, represents a significant and groundbreaking discovery with profound implications for genetic expansion. Romesberg et al. examined the impact of DNA containing 2,6-dimethyl-2H-isoquiniline-1-thione: D5SIC (DS) and 2-methoxy-3-methylnaphthalene: DNAM (DN) UBPs bound to T. aquaticus DNA polymerase (Taq) through crystal structure analysis. Here, we have used polarizable and nonpolarizable classical molecular dynamics (MD) simulations to investigate the structural aspects and stability of Taq in complex with a DNA duplex including a DS-DN pair in the terminal 3' and 5' positions. Our results suggest that the flexibility of UBP-incorporated DNA in the terminal position is arrested by the polymerase, thus preventing fraying and mispairing. Our investigation also reveals that the UBP remains in an intercalated conformation inside the active site, exhibiting two distinct orientations in agreement with experimental findings. Our analysis pinpoints particular residues responsible for favorable interactions with the UBP, with some relying on van der Waals interactions while other on Coulombic forces.
Collapse
Affiliation(s)
- Tanay Debnath
- Department of Physics, University of Texas at Dallas, Richardson, Texas 75080, Dallas, USA.
| | - G Andrés Cisneros
- Department of Physics, University of Texas at Dallas, Richardson, Texas 75080, Dallas, USA.
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, Dallas, USA
| |
Collapse
|
22
|
Debnath T, Cisneros GA. Investigation of dynamical flexibility of D5SIC-DNAM inside DNA duplex in aqueous solution: a systematic classical MD approach. Phys Chem Chem Phys 2024; 26:7435-7445. [PMID: 38353005 PMCID: PMC11080001 DOI: 10.1039/d3cp05572h] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Incorporation of artificial 3rd base pairs (unnatural base pairs, UBPs) has emerged as a fundamental technique in pursuit of expanding the genetic alphabet. 2,6-Dimethyl-2H-isoquiniline-1-thione: D5SIC (DS) and 2-methoxy-3-methylnaphthalene: DNAM (DN), a potential unnatural base pair (UBP) developed by Romesberg and colleagues, has been shown to have remarkable capability for replication within DNA. Crystal structures of a Taq polymerase/double-stranded DNA (ds-DNA) complex containing a DS-DN pair in the 3' terminus showed a parallelly stacked geometry for the pre-insertion, and an intercalated geometry for the post-insertion structure. Unconventional orientations of DS-DN inside a DNA duplex have inspired scientists to investigate the conformational orientations and structural properties of UBP-incorporated DNA. In recent years, computational simulations have been used to investigate the geometry of DS-DN within the DNA duplex; nevertheless, unresolved questions persist owing to inconclusive findings. In this work, we investigate the structural and dynamical properties of DS and DN inside a ds-DNA strand in aqueous solution considering both short and long DNA templates using polarizable, and non-polarizable classical MD simulations. Flexible conformational change of UBP with major populations of Watson-Crick-Franklin (WCF) and three distinct non-Watson-Crick-Franklin (nWCFP1, nWCFP2, nWCFO) conformations through intra and inter-strand flipping have been observed. Our results suggest that a dynamical conformational change leads to the production of diffierent conformational distribution for the systems. Simulations with a short ds-DNA duplex suggest nWCF (P1 and O) as the predominant structures, whereas long ds-DNA duplex simulations indicate almost equal populations of WCF, nWCFP1, nWCFO. DS-DN in the terminal position is found to be more flexible with occasional mispairing and fraying. Overall, these results suggest flexibility and dynamical conformational change of the UBP as well as indicate varied conformational distribution irrespective of starting orientation of the UBP and length og DNA strand.
Collapse
Affiliation(s)
- Tanay Debnath
- Department of Physics, University of Texas at Dallas, Richardson, Texas 75080, Dallas, USA.
| | - G Andrés Cisneros
- Department of Physics, University of Texas at Dallas, Richardson, Texas 75080, Dallas, USA.
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, Dallas, USA
| |
Collapse
|
23
|
Tennakoon M, Thotamune W, Payton JL, Karunarathne A. CaaX-motif-adjacent residues influence G protein gamma (Gγ) prenylation under suboptimal conditions. J Biol Chem 2023; 299:105269. [PMID: 37739036 PMCID: PMC10590752 DOI: 10.1016/j.jbc.2023.105269] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 09/06/2023] [Accepted: 09/09/2023] [Indexed: 09/24/2023] Open
Abstract
Prenylation is an irreversible post-translational modification that supports membrane interactions of proteins involved in various cellular processes, including migration, proliferation, and survival. Dysregulation of prenylation contributes to multiple disorders, including cancers and vascular and neurodegenerative diseases. Prenyltransferases tether isoprenoid lipids to proteins via a thioether linkage during prenylation. Pharmacological inhibition of the lipid synthesis pathway by statins is a therapeutic approach to control hyperlipidemia. Building on our previous finding that statins inhibit membrane association of G protein γ (Gγ) in a subtype-dependent manner, we investigated the molecular reasoning for this differential inhibition. We examined the prenylation of carboxy-terminus (Ct) mutated Gγ in cells exposed to Fluvastatin and prenyl transferase inhibitors and monitored the subcellular localization of fluorescently tagged Gγ subunits and their mutants using live-cell confocal imaging. Reversible optogenetic unmasking-masking of Ct residues was used to probe their contribution to prenylation and membrane interactions of the prenylated proteins. Our findings suggest that specific Ct residues regulate membrane interactions of the Gγ polypeptide, statin sensitivity, and extent of prenylation. Our results also show a few hydrophobic and charged residues at the Ct are crucial determinants of a protein's prenylation ability, especially under suboptimal conditions. Given the cell and tissue-specific expression of different Gγ subtypes, our findings indicate a plausible mechanism allowing for statins to differentially perturb heterotrimeric G protein signaling in cells depending on their Gγ-subtype composition. Our results may also provide molecular reasoning for repurposing statins as Ras oncogene inhibitors and the failure of using prenyltransferase inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Mithila Tennakoon
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri, USA; Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA; Institute for Drug and Biotherapeutic Innovation, Saint Louis University, Saint Louis, Missouri, USA
| | - Waruna Thotamune
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri, USA; Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA; Institute for Drug and Biotherapeutic Innovation, Saint Louis University, Saint Louis, Missouri, USA
| | - John L Payton
- Department of Chemistry, Kenyon College, Gambier, Ohio, USA
| | - Ajith Karunarathne
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri, USA; Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA; Institute for Drug and Biotherapeutic Innovation, Saint Louis University, Saint Louis, Missouri, USA.
| |
Collapse
|
24
|
Tijaro-Bulla S, Nyandwi SP, Cui H. Physiological and engineered tRNA aminoacylation. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1789. [PMID: 37042417 DOI: 10.1002/wrna.1789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/11/2023] [Accepted: 03/21/2023] [Indexed: 04/13/2023]
Abstract
Aminoacyl-tRNA synthetases form the protein family that controls the interpretation of the genetic code, with tRNA aminoacylation being the key chemical step during which an amino acid is assigned to a corresponding sequence of nucleic acids. In consequence, aminoacyl-tRNA synthetases have been studied in their physiological context, in disease states, and as tools for synthetic biology to enable the expansion of the genetic code. Here, we review the fundamentals of aminoacyl-tRNA synthetase biology and classification, with a focus on mammalian cytoplasmic enzymes. We compile evidence that the localization of aminoacyl-tRNA synthetases can be critical in health and disease. In addition, we discuss evidence from synthetic biology which made use of the importance of subcellular localization for efficient manipulation of the protein synthesis machinery. This article is categorized under: RNA Processing Translation > Translation Regulation RNA Processing > tRNA Processing RNA Export and Localization > RNA Localization.
Collapse
Affiliation(s)
| | | | - Haissi Cui
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Davyt M, Bharti N, Ignatova Z. Effect of mRNA/tRNA mutations on translation speed: Implications for human diseases. J Biol Chem 2023; 299:105089. [PMID: 37495112 PMCID: PMC10470029 DOI: 10.1016/j.jbc.2023.105089] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023] Open
Abstract
Recent discoveries establish tRNAs as central regulators of mRNA translation dynamics, and therefore cotranslational folding and function of the encoded protein. The tRNA pool, whose composition and abundance change in a cell- and tissue-dependent manner, is the main factor which determines mRNA translation velocity. In this review, we discuss a group of pathogenic mutations, in the coding sequences of either protein-coding genes or in tRNA genes, that alter mRNA translation dynamics. We also summarize advances in tRNA biology that have uncovered how variations in tRNA levels on account of genetic mutations affect protein folding and function, and thereby contribute to phenotypic diversity in clinical manifestations.
Collapse
Affiliation(s)
- Marcos Davyt
- Institute of Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| | - Nikhil Bharti
- Institute of Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| | - Zoya Ignatova
- Institute of Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany.
| |
Collapse
|
26
|
Tennakoon M, Thotamune W, Payton JL, Karunarathne A. CaaX-motif adjacent residues control G protein prenylation under suboptimal conditions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.04.547731. [PMID: 37461501 PMCID: PMC10349941 DOI: 10.1101/2023.07.04.547731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Prenylation is a universal and irreversible post-translational modification that supports membrane interactions of proteins involved in various cellular processes, including migration, proliferation, and survival. Thus, dysregulation of prenylation contributes to multiple disorders, including cancers, vascular diseases, and neurodegenerative diseases. During prenylation, prenyltransferase enzymes tether metabolically produced isoprenoid lipids to proteins via a thioether linkage. Pharmacological inhibition of the lipid synthesis pathway by statins has long been a therapeutic approach to control hyperlipidemia. Building on our previous finding that statins inhibit membrane association of G protein γ (Gγ) in a subtype-dependent manner, we investigated the molecular reasoning for this differential. We examined the prenylation efficacy of carboxy terminus (Ct) mutated Gγ in cells exposed to Fluvastatin and prenyl transferase inhibitors and monitored the subcellular localization of fluorescently tagged Gγ subunits and their mutants using live-cell confocal imaging. Reversible optogenetic unmasking-masking of Ct residues was used to probe their contribution to the prenylation process and membrane interactions of the prenylated proteins. Our findings suggest that specific Ct residues regulate membrane interactions of the Gγ polypeptide statin sensitivity, and prenylation efficacy. Our results also show that a few hydrophobic and charged residues at the Ct are crucial determinants of a protein's prenylation ability, especially under suboptimal conditions. Given the cell and tissue-specific expression of different Gγ subtypes, our findings explain how and why statins differentially perturb heterotrimeric G protein signaling in specific cells and tissues. Our results may provide molecular reasoning for repurposing statins as Ras oncogene inhibitors and the failure of using prenyltransferase inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Mithila Tennakoon
- Department of Chemistry, Saint Louis University, Saint Louis, MO 63103, USA
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
- Institute for Drug and Biotherapeutic Innovation, Saint Louis University, Saint Louis, MO 63103, USA
| | - Waruna Thotamune
- Department of Chemistry, Saint Louis University, Saint Louis, MO 63103, USA
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
- Institute for Drug and Biotherapeutic Innovation, Saint Louis University, Saint Louis, MO 63103, USA
| | - John L. Payton
- Department of Chemistry, Kenyon College, Gambier, Ohio 43022, USA
| | - Ajith Karunarathne
- Department of Chemistry, Saint Louis University, Saint Louis, MO 63103, USA
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
- Institute for Drug and Biotherapeutic Innovation, Saint Louis University, Saint Louis, MO 63103, USA
| |
Collapse
|
27
|
Fricke R, Swenson CV, Roe LT, Hamlish NX, Shah B, Zhang Z, Ficaretta E, Ad O, Smaga S, Gee CL, Chatterjee A, Schepartz A. Expanding the substrate scope of pyrrolysyl-transfer RNA synthetase enzymes to include non-α-amino acids in vitro and in vivo. Nat Chem 2023; 15:960-971. [PMID: 37264106 PMCID: PMC10322718 DOI: 10.1038/s41557-023-01224-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/28/2023] [Indexed: 06/03/2023]
Abstract
The absence of orthogonal aminoacyl-transfer RNA (tRNA) synthetases that accept non-L-α-amino acids is a primary bottleneck hindering the in vivo translation of sequence-defined hetero-oligomers and biomaterials. Here we report that pyrrolysyl-tRNA synthetase (PylRS) and certain PylRS variants accept α-hydroxy, α-thio and N-formyl-L-α-amino acids, as well as α-carboxy acid monomers that are precursors to polyketide natural products. These monomers are accommodated and accepted by the translation apparatus in vitro; those with reactive nucleophiles are incorporated into proteins in vivo. High-resolution structural analysis of the complex formed between one PylRS enzyme and a m-substituted 2-benzylmalonic acid derivative revealed an active site that discriminates prochiral carboxylates and accommodates the large size and distinct electrostatics of an α-carboxy substituent. This work emphasizes the potential of PylRS-derived enzymes for acylating tRNA with monomers whose α-substituent diverges substantially from the α-amine of proteinogenic amino acids. These enzymes or derivatives thereof could synergize with natural or evolved ribosomes and/or translation factors to generate diverse sequence-defined non-protein heteropolymers.
Collapse
Affiliation(s)
- Riley Fricke
- Department of Chemistry, University of California, Berkeley, CA, USA
- Center for Genetically Encoded Materials, University of California, Berkeley, CA, USA
| | - Cameron V Swenson
- Department of Chemistry, University of California, Berkeley, CA, USA
- Center for Genetically Encoded Materials, University of California, Berkeley, CA, USA
| | - Leah Tang Roe
- Department of Chemistry, University of California, Berkeley, CA, USA
- Center for Genetically Encoded Materials, University of California, Berkeley, CA, USA
| | - Noah Xue Hamlish
- Center for Genetically Encoded Materials, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Bhavana Shah
- Process Development, Amgen, Thousand Oaks, CA, USA
| | | | - Elise Ficaretta
- Center for Genetically Encoded Materials, University of California, Berkeley, CA, USA
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Omer Ad
- Department of Chemistry, Yale University, New Haven, CT, USA
| | - Sarah Smaga
- Department of Chemistry, University of California, Berkeley, CA, USA
- Center for Genetically Encoded Materials, University of California, Berkeley, CA, USA
| | - Christine L Gee
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, USA
| | - Abhishek Chatterjee
- Center for Genetically Encoded Materials, University of California, Berkeley, CA, USA
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Alanna Schepartz
- Department of Chemistry, University of California, Berkeley, CA, USA.
- Center for Genetically Encoded Materials, University of California, Berkeley, CA, USA.
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, USA.
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA, USA.
| |
Collapse
|
28
|
Ji Y, Liu D, Zhu H, Bao L, Chang R, Gao X, Yin J. Unstructured Polypeptides as a Versatile Drug Delivery Technology. Acta Biomater 2023; 164:74-93. [PMID: 37075961 DOI: 10.1016/j.actbio.2023.04.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/23/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023]
Abstract
Although polyethylene glycol (PEG), or "PEGylation" has become a widely applied approach for improving the efficiency of drug delivery, the immunogenicity and non-biodegradability of this synthetic polymer have prompted an evident need for alternatives. To overcome these caveats and to mimic PEG -or other natural or synthetic polymers- for the purpose of drug half-life extension, unstructured polypeptides are designed. Due to their tunable length, biodegradability, low immunogenicity and easy production, unstructured polypeptides have the potential to replace PEG as the preferred technology for therapeutic protein/peptide delivery. This review provides an overview of the evolution of unstructured polypeptides, starting from natural polypeptides to engineered polypeptides and discusses their characteristics. Then, it is described that unstructured polypeptides have been successfully applied to numerous drugs, including peptides, proteins, antibody fragments, and nanocarriers, for half-life extension. Innovative applications of unstructured peptides as releasable masks, multimolecular adaptors and intracellular delivery carriers are also discussed. Finally, challenges and future perspectives of this promising field are briefly presented. STATEMENT OF SIGNIFICANCE: : Polypeptide fusion technology simulating PEGylation has become an important topic for the development of long-circulating peptide or protein drugs without reduced activity, complex processes, and kidney injury caused by PEG modification. Here we provide a detailed and in-depth review of the recent advances in unstructured polypeptides. In addition to the application of enhanced pharmacokinetic performance, emphasis is placed on polypeptides as scaffolders for the delivery of multiple drugs, and on the preparation of reasonably designed polypeptides to manipulate the performance of proteins and peptides. This review will provide insight into future application of polypeptides in peptide or protein drug development and the design of novel functional polypeptides.
Collapse
Affiliation(s)
- Yue Ji
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Dingkang Liu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Haichao Zhu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Lichen Bao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 210009, China
| | - Ruilong Chang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Jun Yin
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
29
|
Słota D, Piętak K, Jampilek J, Sobczak-Kupiec A. Polymeric and Composite Carriers of Protein and Non-Protein Biomolecules for Application in Bone Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2023; 16:2235. [PMID: 36984115 PMCID: PMC10059071 DOI: 10.3390/ma16062235] [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: 01/30/2023] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 06/18/2023]
Abstract
Conventional intake of drugs and active substances is most often based on oral intake of an appropriate dose to achieve the desired effect in the affected area or source of pain. In this case, controlling their distribution in the body is difficult, as the substance also reaches other tissues. This phenomenon results in the occurrence of side effects and the need to increase the concentration of the therapeutic substance to ensure it has the desired effect. The scientific field of tissue engineering proposes a solution to this problem, which creates the possibility of designing intelligent systems for delivering active substances precisely to the site of disease conversion. The following review discusses significant current research strategies as well as examples of polymeric and composite carriers for protein and non-protein biomolecules designed for bone tissue regeneration.
Collapse
Affiliation(s)
- Dagmara Słota
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| | - Karina Piętak
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| | - Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
- Department of Chemical Biology, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 783 71 Olomouc, Czech Republic
| | - Agnieszka Sobczak-Kupiec
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| |
Collapse
|
30
|
Lee K, Willi JA, Cho N, Kim I, Jewett MC, Lee J. Cell-free Biosynthesis of Peptidomimetics. BIOTECHNOL BIOPROC E 2023; 28:1-17. [PMID: 36778039 PMCID: PMC9896473 DOI: 10.1007/s12257-022-0268-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/16/2022] [Accepted: 11/13/2022] [Indexed: 02/05/2023]
Abstract
A wide variety of peptidomimetics (peptide analogs) possessing innovative biological functions have been brought forth as therapeutic candidates through cell-free protein synthesis (CFPS) systems. A key feature of these peptidomimetic drugs is the use of non-canonical amino acid building blocks with diverse biochemical properties that expand functional diversity. Here, we summarize recent technologies leveraging CFPS platforms to expand the reach of peptidomimetics drugs. We also offer perspectives on engineering the translational machinery that may open new opportunities for expanding genetically encoded chemistry to transform drug discovery practice beyond traditional boundaries.
Collapse
Affiliation(s)
- Kanghun Lee
- School of Interdisciplinary Bioscience and Bioengineering (I-Bio), Pohang University of Science and Technology (POSTECH), Pohang, 37673 Korea
| | - Jessica A. Willi
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208 USA
| | - Namjin Cho
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Korea
| | - Inseon Kim
- School of Interdisciplinary Bioscience and Bioengineering (I-Bio), Pohang University of Science and Technology (POSTECH), Pohang, 37673 Korea
| | - Michael C. Jewett
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208 USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL 60208 USA
| | - Joongoo Lee
- School of Interdisciplinary Bioscience and Bioengineering (I-Bio), Pohang University of Science and Technology (POSTECH), Pohang, 37673 Korea
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Korea
| |
Collapse
|
31
|
Yeung YW, Chan CM, Chen YT, Chan M, Luo M, Gao X, Du B, Yu WY. Cu-Catalyzed Cross-Electrophilic Coupling of α-Diazoesters with O-Benzoyl Hydroxylamines for the Synthesis of Unnatural N-Alkyl α-Amino Acid Derivatives. Org Lett 2023; 25:619-623. [PMID: 36692243 DOI: 10.1021/acs.orglett.2c04161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We describe a Cu-catalyzed cross-electrophilic coupling reaction for synthesizing α-amino acid derivatives from α-diazoesters with O-benzoyl hydroxylamines with Cu(OAc)2 as the catalyst and polymethylhydrosilane (PMHS) as the hydride reagent. Excellent functional group compatibilities were demonstrated. With ethyl 2-diazo-3-oxobutanoate as the precursor, a Cu-acetoacetate complex has been characterized by ESI-MS analysis. Results from the radical trap experiments are consistent with the intermediacy of nitrogen-centered radicals. This strategy offers a simple and inexpensive synthesis of α-amino acid derivatives.
Collapse
Affiliation(s)
- Yiu-Wai Yeung
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Chun-Ming Chan
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Yu-Ting Chen
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Marco Chan
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Mingyu Luo
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Xin Gao
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210098, China
| | - Bingnan Du
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210098, China
| | - Wing-Yiu Yu
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| |
Collapse
|
32
|
Thoma B, Powner MW. Selective Synthesis of Lysine Peptides and the Prebiotically Plausible Synthesis of Catalytically Active Diaminopropionic Acid Peptide Nitriles in Water. J Am Chem Soc 2023; 145:3121-3130. [PMID: 36700882 PMCID: PMC9912261 DOI: 10.1021/jacs.2c12497] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Why life encodes specific proteinogenic amino acids remains an unsolved problem, but a non-enzymatic synthesis that recapitulates biology's universal strategy of stepwise N-to-C terminal peptide growth may hold the key to this selection. Lysine is an important proteinogenic amino acid that, despite its essential structural, catalytic, and functional roles in biochemistry, has widely been assumed to be a late addition to the genetic code. Here, we demonstrate that lysine thioacids undergo coupling with aminonitriles in neutral water to afford peptides in near-quantitative yield, whereas non-proteinogenic lysine homologues, ornithine, and diaminobutyric acid cannot form peptides due to rapid and quantitative cyclization that irreversibly blocks peptide synthesis. We demonstrate for the first time that ornithine lactamization provides an absolute differentiation of lysine and ornithine during (non-enzymatic) N-to-C-terminal peptide ligation. We additionally demonstrate that the shortest lysine homologue, diaminopropionic acid, undergoes effective peptide ligation. This prompted us to discover a high-yielding prebiotically plausible synthesis of the diaminopropionic acid residue, by peptide nitrile modification, through the addition of ammonia to a dehydroalanine nitrile. With this synthesis in hand, we then discovered that the low basicity of diaminopropionyl residues promotes effective, biomimetic, imine catalysis in neutral water. Our results suggest diaminopropionic acid, synthesized by peptide nitrile modification, can replace or augment lysine residues during early evolution but that lysine's electronically isolated sidechain amine likely provides an evolutionary advantage for coupling and coding as a preformed monomer in monomer-by-monomer peptide translation.
Collapse
|
33
|
A strategy for screening and identification of new amino acid-conjugated bile acids with high coverage by liquid chromatography-mass spectrometry. Anal Chim Acta 2023; 1239:340691. [PMID: 36628759 DOI: 10.1016/j.aca.2022.340691] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022]
Abstract
Bile acids (BAs) are a class of vital gut microbiota-host cometabolites, and they play an important role in maintaining gut microbiota-host metabolic homeostasis. Very recently, a new mechanism of BA anabolic metabolism mediated by gut microbiota (BA-amino acid conjugation) has been revealed, which provides a perspective for the research on BA metabolism and gut metabolome. In this study, we established a polarity-switching multiple reaction monitoring mass spectrometry-based screening method to mine amino acid-conjugated bile acids (AA-BAs) derived from host-gut microbiota co-metabolism. In addition, a retention time-based annotation strategy was further proposed to identify the AA-BA isomers and epimers. Using the developed methods, we successfully screened 118 AA-BA conjugates from mouse and human feces, 28 of them were confirmed by standards, and 62 putatively identified based on their predicted retention times. Moreover, we observed that the levels of most AA-BAs were significantly downregulated in the feces of chronic sleep deprivation mice, suggesting that the AA-BA metabolism was closely related to the physiological state of the host.
Collapse
|
34
|
Santos FB, Del-Bem LE. The Evolution of tRNA Copy Number and Repertoire in Cellular Life. Genes (Basel) 2022; 14:27. [PMID: 36672768 PMCID: PMC9858662 DOI: 10.3390/genes14010027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
tRNAs are universal decoders that bridge the gap between transcriptome and proteome. They can also be processed into small RNA fragments with regulatory functions. In this work, we show that tRNA copy number is largely controlled by genome size in all cellular organisms, in contrast to what is observed for protein-coding genes that stop expanding between ~20,000 and ~35,000 loci per haploid genome in eukaryotes, regardless of genome size. Our analyses indicate that after the bacteria/archaea split, the tRNA gene pool experienced the evolution of increased anticodon diversity in the archaeal lineage, along with a tRNA gene size increase and mature tRNA size decrease. The evolution and diversification of eukaryotes from archaeal ancestors involved further expansion of the tRNA anticodon repertoire, additional increase in tRNA gene size and decrease in mature tRNA length, along with an explosion of the tRNA gene copy number that emerged coupled with accelerated genome size expansion. Our findings support the notion that macroscopic eukaryotes with a high diversity of cell types, such as land plants and vertebrates, independently evolved a high diversity of tRNA anticodons along with high gene redundancy caused by the expansion of the tRNA copy number. The results presented here suggest that the evolution of tRNA genes played important roles in the early split between bacteria and archaea, and in eukaryogenesis and the later emergence of complex eukaryotes, with potential implications in protein translation and gene regulation through tRNA-derived RNA fragments.
Collapse
Affiliation(s)
- Fenícia Brito Santos
- Del-Bem Lab, Department of Botany, Institute of Biological Sciences (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil
- Graduate Program in Bioinformatics, Institute of Biological Sciences (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil
| | - Luiz-Eduardo Del-Bem
- Del-Bem Lab, Department of Botany, Institute of Biological Sciences (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil
| |
Collapse
|
35
|
Fages‐Lartaud M, Hundvin K, Hohmann‐Marriott MF. Mechanisms governing codon usage bias and the implications for protein expression in the chloroplast of Chlamydomonas reinhardtii. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2022; 112:919-945. [PMID: 36071273 PMCID: PMC9828097 DOI: 10.1111/tpj.15970] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 05/30/2023]
Abstract
Chloroplasts possess a considerably reduced genome that is decoded via an almost minimal set of tRNAs. These features make an excellent platform for gaining insights into fundamental mechanisms that govern protein expression. Here, we present a comprehensive and revised perspective of the mechanisms that drive codon selection in the chloroplast of Chlamydomonas reinhardtii and the functional consequences for protein expression. In order to extract this information, we applied several codon usage descriptors to genes with different expression levels. We show that highly expressed genes strongly favor translationally optimal codons, while genes with lower functional importance are rather affected by directional mutational bias. We demonstrate that codon optimality can be deduced from codon-anticodon pairing affinity and, for a small number of amino acids (leucine, arginine, serine, and isoleucine), tRNA concentrations. Finally, we review, analyze, and expand on the impact of codon usage on protein yield, secondary structures of mRNA, translation initiation and termination, and amino acid composition of proteins, as well as cotranslational protein folding. The comprehensive analysis of codon choice provides crucial insights into heterologous gene expression in the chloroplast of C. reinhardtii, which may also be applicable to other chloroplast-containing organisms and bacteria.
Collapse
Affiliation(s)
- Maxime Fages‐Lartaud
- Department of BiotechnologyNorwegian University of Science and TechnologyTrondheimN‐7491Norway
| | - Kristoffer Hundvin
- Department of BiotechnologyNorwegian University of Science and TechnologyTrondheimN‐7491Norway
| | | |
Collapse
|
36
|
Chen Y, Jin S, Zhang M, Hu Y, Wu KL, Chung A, Wang S, Tian Z, Wang Y, Wolynes PG, Xiao H. Unleashing the potential of noncanonical amino acid biosynthesis to create cells with precision tyrosine sulfation. Nat Commun 2022; 13:5434. [PMID: 36114189 PMCID: PMC9481576 DOI: 10.1038/s41467-022-33111-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 09/01/2022] [Indexed: 01/31/2023] Open
Abstract
Despite the great promise of genetic code expansion technology to modulate structures and functions of proteins, external addition of ncAAs is required in most cases and it often limits the utility of genetic code expansion technology, especially to noncanonical amino acids (ncAAs) with poor membrane internalization. Here, we report the creation of autonomous cells, both prokaryotic and eukaryotic, with the ability to biosynthesize and genetically encode sulfotyrosine (sTyr), an important protein post-translational modification with low membrane permeability. These engineered cells can produce site-specifically sulfated proteins at a higher yield than cells fed exogenously with the highest level of sTyr reported in the literature. We use these autonomous cells to prepare highly potent thrombin inhibitors with site-specific sulfation. By enhancing ncAA incorporation efficiency, this added ability of cells to biosynthesize ncAAs and genetically incorporate them into proteins greatly extends the utility of genetic code expansion methods.
Collapse
Affiliation(s)
- Yuda Chen
- grid.21940.3e0000 0004 1936 8278Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005 USA
| | - Shikai Jin
- grid.21940.3e0000 0004 1936 8278Center for Theoretical Biological Physics, Rice University, 6100 Main Street, Houston, TX 77005 USA ,grid.21940.3e0000 0004 1936 8278Department of Biosciences, Rice University, 6100 Main Street, Houston, TX 77005 USA
| | - Mengxi Zhang
- grid.21940.3e0000 0004 1936 8278Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005 USA
| | - Yu Hu
- grid.21940.3e0000 0004 1936 8278Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005 USA
| | - Kuan-Lin Wu
- grid.21940.3e0000 0004 1936 8278Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005 USA
| | - Anna Chung
- grid.21940.3e0000 0004 1936 8278Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005 USA
| | - Shichao Wang
- grid.21940.3e0000 0004 1936 8278Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005 USA
| | - Zeru Tian
- grid.21940.3e0000 0004 1936 8278Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005 USA
| | - Yixian Wang
- grid.21940.3e0000 0004 1936 8278Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005 USA
| | - Peter G. Wolynes
- grid.21940.3e0000 0004 1936 8278Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005 USA ,grid.21940.3e0000 0004 1936 8278Center for Theoretical Biological Physics, Rice University, 6100 Main Street, Houston, TX 77005 USA ,grid.21940.3e0000 0004 1936 8278Department of Biosciences, Rice University, 6100 Main Street, Houston, TX 77005 USA ,grid.21940.3e0000 0004 1936 8278Department of Physics, Rice University, 6100 Main Street, Houston, TX 77005 USA
| | - Han Xiao
- grid.21940.3e0000 0004 1936 8278Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005 USA ,grid.21940.3e0000 0004 1936 8278Department of Biosciences, Rice University, 6100 Main Street, Houston, TX 77005 USA ,grid.21940.3e0000 0004 1936 8278Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX 77005 USA
| |
Collapse
|
37
|
Selenoprotein: Potential Player in Redox Regulation in Chlamydomonas reinhardtii. Antioxidants (Basel) 2022; 11:antiox11081630. [PMID: 36009349 PMCID: PMC9404770 DOI: 10.3390/antiox11081630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/22/2022] Open
Abstract
Selenium (Se) is an essential micro-element for many organisms, including Chlamydomonas reinhardtii, and is required in trace amounts. It is obtained from the 21st amino acid selenocysteine (Sec, U), genetically encoded by the UGA codon. Proteins containing Sec are known as selenoproteins. In eukaryotes, selenoproteins are present in animals and algae, whereas fungi and higher plants lack them. The human genome contains 25 selenoproteins, most of which are involved in antioxidant defense activity, redox regulation, and redox signaling. In algae, 42 selenoprotein families were identified using various bioinformatics approaches, out of which C. reinhardtii is known to have 10 selenoprotein genes. However, the role of selenoproteins in Chlamydomonas is yet to be reported. Chlamydomonas selenoproteins contain conserved domains such as CVNVGC and GCUG, in the case of thioredoxin reductase, and CXXU in other selenoproteins. Interestingly, Sec amino acid residue is present in a catalytically active domain in Chlamydomonas selenoproteins, similar to human selenoproteins. Based on catalytical active sites and conserved domains present in Chlamydomonas selenoproteins, we suggest that Chlamydomonas selenoproteins could have a role in redox regulation and defense by acting as antioxidants in various physiological conditions.
Collapse
|
38
|
Deciphering the conformational landscape of few selected aromatic noncoded amino acids (NCAAs) for applications in rational design of peptide therapeutics. Amino Acids 2022; 54:1183-1202. [PMID: 35723743 PMCID: PMC9207436 DOI: 10.1007/s00726-022-03175-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 05/23/2022] [Indexed: 11/01/2022]
Abstract
Amino acids are the essential building blocks of both synthetic and natural peptides, which are crucial for biological functions and also important as biological probes for mapping the complex protein-protein interactions (PPIs) in both prokaryotic and eukaryotic systems. Mapping the PPIs through the chemical biology approach provides pharmacologically relevant peptides, which can have agonistic or antagonistic effects on the targeted biological systems. It is evidenced that ≥ 60 peptide-based drugs have been approved by the US-FDA so far, and the number will improve further in the foreseeable future, as ≥ 140 peptides are currently in clinical trials. However, natural peptides often require fine-tuning of their pharmacological properties by strategically replacing the αL-amino acids of the peptides with non-coded amino acids (NCAA), for which codons are absent in the genetic code for biosynthesis of proteins, prior to their applications as therapeutics. Considering the diverse repertoire of the NCAAs, the conformational space of many NCAAs is yet to be explored systematically in the context of the rational design of therapeutic peptides. The current study deciphers the conformational landscape of a few such Cα-substituted aromatic NCAAs (Ing: 2-indanyl-L-Glycine; Bpa: 4-benzoyl-L-phenylalanine; Aic: 2-aminoindane-2-carboxylic acid) both in the context of tripeptides and model synthetic peptide sequences, using alanine (Ala) and proline (Pro) as the reference. The combined data obtained from the computational and biophysical studies indicate the general success of this approach, which can be exploited further to rationally design optimized peptide sequences of unusual architecture with potent antimicrobial, antiviral, gluco-regulatory, immunomodulatory, and anti-inflammatory activities.
Collapse
|
39
|
Kimoto M, Hirao I. Genetic Code Engineering by Natural and Unnatural Base Pair Systems for the Site-Specific Incorporation of Non-Standard Amino Acids Into Proteins. Front Mol Biosci 2022; 9:851646. [PMID: 35685243 PMCID: PMC9171071 DOI: 10.3389/fmolb.2022.851646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/25/2022] [Indexed: 12/21/2022] Open
Abstract
Amino acid sequences of proteins are encoded in nucleic acids composed of four letters, A, G, C, and T(U). However, this four-letter alphabet coding system limits further functionalities of proteins by the twenty letters of amino acids. If we expand the genetic code or develop alternative codes, we could create novel biological systems and biotechnologies by the site-specific incorporation of non-standard amino acids (or unnatural amino acids, unAAs) into proteins. To this end, new codons and their complementary anticodons are required for unAAs. In this review, we introduce the current status of methods to incorporate new amino acids into proteins by in vitro and in vivo translation systems, by focusing on the creation of new codon-anticodon interactions, including unnatural base pair systems for genetic alphabet expansion.
Collapse
Affiliation(s)
| | - Ichiro Hirao
- *Correspondence: Michiko Kimoto, ; Ichiro Hirao,
| |
Collapse
|
40
|
Perez JG, Carlson ED, Weisser O, Kofman C, Seki K, Des Soye BJ, Karim AS, Jewett MC. Improving genomically recoded Escherichia coli to produce proteins containing non-canonical amino acids. Biotechnol J 2022; 17:e2100330. [PMID: 34894206 DOI: 10.1002/biot.202100330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022]
Abstract
A genomically recoded Escherichia coli strain that lacks all amber codons and release factor 1 (C321.∆A) enables efficient genetic encoding of chemically diverse non-canonical amino acids (ncAAs) into proteins. While C321.∆A has opened new opportunities in chemical and synthetic biology, this strain has not been optimized for protein production, limiting its utility in widespread industrial and academic applications. To address this limitation, the construction of a series of genomically recoded organisms that are optimized for cellular protein production is described. It is demonstrated that the functional deactivation of nucleases (e.g., rne, endA) and proteases (e.g., lon) increases production of wild-type superfolder green fluorescent protein (sfGFP) and sfGFP containing two ncAAs up to ≈5-fold. Additionally, a genomic IPTG-inducible T7 RNA polymerase (T7RNAP) cassette into these strains is introduced. Using an optimized platform, the ability to introduce two identical N6 -(propargyloxycarbonyl)-L -Lysine residues site specifically into sfGFP with a 17-fold improvement in production relative to the parent strain is demonstrated. The authors envision that their library of organisms will provide the community with multiple options for increased expression of proteins with new and diverse chemistries.
Collapse
Affiliation(s)
- Jessica G Perez
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA
| | - Erik D Carlson
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA
| | - Oliver Weisser
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA
| | - Camila Kofman
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA
| | - Kosuke Seki
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA
| | - Benjamin J Des Soye
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA
| | - Ashty S Karim
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA
| | - Michael C Jewett
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA
- Center for Synthetic Biology, Northwestern University, Evanston, Illinois, USA
- Simpson Querrey Institute, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
41
|
Freeland S. Undefining life's biochemistry: implications for abiogenesis. J R Soc Interface 2022; 19:20210814. [PMID: 35193384 PMCID: PMC8867283 DOI: 10.1098/rsif.2021.0814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/19/2022] [Indexed: 12/22/2022] Open
Abstract
In the mid-twentieth century, multiple Nobel Prizes rewarded discoveries of a seemingly universal set of molecules and interactions that collectively defined the chemical basis for life. Twenty-first-century science knows that every detail of this Central Dogma of Molecular Biology can vary through either biological evolution, human engineering (synthetic biology) or both. Clearly the material, molecular basis of replicating, evolving entities can be different. There is far less clarity yet for what constitutes this set of possibilities. One approach to better understand the limits and scope of moving beyond life's central dogma comes from those who study life's origins. RNA, proteins and the genetic code that binds them each look like products of natural selection. This raises the question of what step(s) preceded these particular components? Answers here will clarify whether any discrete point in time or biochemical evolution will objectively merit the label of life's origin, or whether life unfolds seamlessly from the non-living universe.
Collapse
Affiliation(s)
- Stephen Freeland
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| |
Collapse
|
42
|
Eugenio FA, van Milgen J, Duperray J, Sergheraert R, Le Floc'h N. Feeding intact proteins, peptides, or free amino acids to monogastric farm animals. Amino Acids 2022; 54:157-168. [PMID: 35106634 DOI: 10.1007/s00726-021-03118-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022]
Abstract
For terrestrial farm animals, intact protein sources like soybean meal have been the main ingredients providing the required amino acids (AA) to sustain life. However, in recent years, the availability of hydrolysed protein sources and free AA has led to the use of other forms of AA to feed farm animals. The advent of using these new forms is especially important to reduce the negative environmental impacts of animal production because these new forms allow reducing the dietary crude protein content and provide more digestible materials. However, the form in which dietary AA are provided can have an effect on the dynamics of nutrient availability for protein deposition and tissue growth including the efficiency of nutrient utilization. In this literature review, the use of different forms of AA in animal diets is explored, and their differences in digestion and absorption rates are focused on. These differences affect the postprandial plasma appearance of AA, which can have metabolic consequences, like greater insulin response when free AA or hydrolysates instead of intact proteins are fed, which can have a profound effect on metabolism and growth performance. Nevertheless, the use and application of the different AA forms in animal diets are important to achieve a more sustainable and efficient animal production system in the future, as they allow for a more precise diet formulation and reduced negative environmental impact. It is, therefore, important to differentiate the physiological and metabolic effects of different forms of AA to maximize their nutritional value in animal diets.
Collapse
Affiliation(s)
- F A Eugenio
- PEGASE, INRAE, Institut Agro, 35590, Saint Gilles, France
- BCF Life Sciences, Boisel, 56140, Pleucadeuc, France
| | - J van Milgen
- PEGASE, INRAE, Institut Agro, 35590, Saint Gilles, France
| | - J Duperray
- BCF Life Sciences, Boisel, 56140, Pleucadeuc, France
| | - R Sergheraert
- BCF Life Sciences, Boisel, 56140, Pleucadeuc, France
| | - N Le Floc'h
- PEGASE, INRAE, Institut Agro, 35590, Saint Gilles, France.
| |
Collapse
|
43
|
Zhang Z, Liao D, Ma Y, Jia B, Yuan Y. Orthogonality of Redesigned
tRNA
Molecules with Three Stop Codons. CHINESE J CHEM 2022. [DOI: 10.1002/cjoc.202100759] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Zhao‐Yang Zhang
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology Tianjin University Tianjin 300072 China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) Tianjin University Tianjin 300072 China
| | - Dan‐Ni Liao
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology Tianjin University Tianjin 300072 China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) Tianjin University Tianjin 300072 China
| | - Yu‐Xin Ma
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology Tianjin University Tianjin 300072 China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) Tianjin University Tianjin 300072 China
| | - Bin Jia
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology Tianjin University Tianjin 300072 China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) Tianjin University Tianjin 300072 China
| | - Ying‐Jin Yuan
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology Tianjin University Tianjin 300072 China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) Tianjin University Tianjin 300072 China
| |
Collapse
|
44
|
Lateef OM, Akintubosun MO, Olaoba OT, Samson SO, Adamczyk M. Making Sense of "Nonsense" and More: Challenges and Opportunities in the Genetic Code Expansion, in the World of tRNA Modifications. Int J Mol Sci 2022; 23:938. [PMID: 35055121 PMCID: PMC8779196 DOI: 10.3390/ijms23020938] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 01/09/2023] Open
Abstract
The evolutional development of the RNA translation process that leads to protein synthesis based on naturally occurring amino acids has its continuation via synthetic biology, the so-called rational bioengineering. Genetic code expansion (GCE) explores beyond the natural translational processes to further enhance the structural properties and augment the functionality of a wide range of proteins. Prokaryotic and eukaryotic ribosomal machinery have been proven to accept engineered tRNAs from orthogonal organisms to efficiently incorporate noncanonical amino acids (ncAAs) with rationally designed side chains. These side chains can be reactive or functional groups, which can be extensively utilized in biochemical, biophysical, and cellular studies. Genetic code extension offers the contingency of introducing more than one ncAA into protein through frameshift suppression, multi-site-specific incorporation of ncAAs, thereby increasing the vast number of possible applications. However, different mediating factors reduce the yield and efficiency of ncAA incorporation into synthetic proteins. In this review, we comment on the recent advancements in genetic code expansion to signify the relevance of systems biology in improving ncAA incorporation efficiency. We discuss the emerging impact of tRNA modifications and metabolism in protein design. We also provide examples of the latest successful accomplishments in synthetic protein therapeutics and show how codon expansion has been employed in various scientific and biotechnological applications.
Collapse
Affiliation(s)
- Olubodun Michael Lateef
- Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (O.M.L.); (M.O.A.); (S.O.S.)
| | | | - Olamide Tosin Olaoba
- Laboratory of Functional and Structural Biochemistry, Federal University of Sao Carlos, Sao Carlos 13565-905, SP, Brazil;
| | - Sunday Ocholi Samson
- Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (O.M.L.); (M.O.A.); (S.O.S.)
| | - Malgorzata Adamczyk
- Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (O.M.L.); (M.O.A.); (S.O.S.)
| |
Collapse
|
45
|
NAKAMURA E, KOBAYASHI K, TANAKA R, KUNIHIRO T, KITAGAWA H, POTISZIL C, OTA T, SAKAGUCHI C, YAMANAKA M, RATNAYAKE DM, TRIPATHI H, KUMAR R, AVRAMESCU ML, TSUCHIDA H, YACHI Y, MIURA H, ABE M, FUKAI R, FURUYA S, HATAKEDA K, HAYASHI T, HITOMI Y, KUMAGAI K, MIYAZAKI A, NAKATO A, NISHIMURA M, OKADA T, SOEJIMA H, SUGITA S, SUZUKI A, USUI T, YADA T, YAMAMOTO D, YOGATA K, YOSHITAKE M, ARAKAWA M, FUJII A, HAYAKAWA M, HIRATA N, HIRATA N, HONDA R, HONDA C, HOSODA S, IIJIMA YI, IKEDA H, ISHIGURO M, ISHIHARA Y, IWATA T, KAWAHARA K, KIKUCHI S, KITAZATO K, MATSUMOTO K, MATSUOKA M, MICHIKAMI T, MIMASU Y, MIURA A, MOROTA T, NAKAZAWA S, NAMIKI N, NODA H, NOGUCHI R, OGAWA N, OGAWA K, OKAMOTO C, ONO G, OZAKI M, SAIKI T, SAKATANI N, SAWADA H, SENSHU H, SHIMAKI Y, SHIRAI K, TAKEI Y, TAKEUCHI H, TANAKA S, TATSUMI E, TERUI F, TSUKIZAKI R, WADA K, YAMADA M, YAMADA T, YAMAMOTO Y, YANO H, YOKOTA Y, YOSHIHARA K, YOSHIKAWA M, YOSHIKAWA K, FUJIMOTO M, WATANABE SI, TSUDA Y. On the origin and evolution of the asteroid Ryugu: A comprehensive geochemical perspective. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2022; 98:227-282. [PMID: 35691845 PMCID: PMC9246647 DOI: 10.2183/pjab.98.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/06/2022] [Indexed: 05/28/2023]
Abstract
Presented here are the observations and interpretations from a comprehensive analysis of 16 representative particles returned from the C-type asteroid Ryugu by the Hayabusa2 mission. On average Ryugu particles consist of 50% phyllosilicate matrix, 41% porosity and 9% minor phases, including organic matter. The abundances of 70 elements from the particles are in close agreement with those of CI chondrites. Bulk Ryugu particles show higher δ18O, Δ17O, and ε54Cr values than CI chondrites. As such, Ryugu sampled the most primitive and least-thermally processed protosolar nebula reservoirs. Such a finding is consistent with multi-scale H-C-N isotopic compositions that are compatible with an origin for Ryugu organic matter within both the protosolar nebula and the interstellar medium. The analytical data obtained here, suggests that complex soluble organic matter formed during aqueous alteration on the Ryugu progenitor planetesimal (several 10's of km), <2.6 Myr after CAI formation. Subsequently, the Ryugu progenitor planetesimal was fragmented and evolved into the current asteroid Ryugu through sublimation.
Collapse
Affiliation(s)
- Eizo NAKAMURA
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Katsura KOBAYASHI
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Ryoji TANAKA
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Tak KUNIHIRO
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Hiroshi KITAGAWA
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Christian POTISZIL
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Tsutomu OTA
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Chie SAKAGUCHI
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Masahiro YAMANAKA
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Dilan M. RATNAYAKE
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Havishk TRIPATHI
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Rahul KUMAR
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Maya-Liliana AVRAMESCU
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Hidehisa TSUCHIDA
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Yusuke YACHI
- The Pheasant Memorial Laboratory for Geochemistry and Cosmochemistry, Institute for Planetary Materials, Okayama University, Misasa, Tottori, Japan
| | - Hitoshi MIURA
- Department of Information and Basic Science, Nagoya City University, Nagoya, Aichi, Japan
| | - Masanao ABE
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- The Graduate University for Advanced Studies (SOKENDAI), Hayama, Kanagawa, Japan
| | - Ryota FUKAI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Shizuho FURUYA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Kentaro HATAKEDA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Tasuku HAYASHI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Yuya HITOMI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- Marine Works Japan, Ltd., Yokosuka, Kanagawa, Japan
| | - Kazuya KUMAGAI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- Marine Works Japan, Ltd., Yokosuka, Kanagawa, Japan
| | - Akiko MIYAZAKI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Aiko NAKATO
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Masahiro NISHIMURA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Tatsuaki OKADA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Hiromichi SOEJIMA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- Marine Works Japan, Ltd., Yokosuka, Kanagawa, Japan
| | - Seiji SUGITA
- Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Planetary Exploration Research Center (PERC), Chiba Institute of Technology, Narashino, Chiba, Japan
| | - Ayako SUZUKI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- Marine Works Japan, Ltd., Yokosuka, Kanagawa, Japan
| | - Tomohiro USUI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Toru YADA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Daiki YAMAMOTO
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Kasumi YOGATA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Miwa YOSHITAKE
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | | | - Atsushi FUJII
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Masahiko HAYAKAWA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Naoyuki HIRATA
- Graduate School of Science, Kobe University, Kobe, Hyogo, Japan
| | - Naru HIRATA
- Faculty of Computer Science and Engineering, The University of Aizu, Aizu-Wakamatsu, Fukushima, Japan
| | - Rie HONDA
- Faculty of Science and Technology, Kochi University, Kochi, Japan
| | - Chikatoshi HONDA
- Faculty of Computer Science and Engineering, The University of Aizu, Aizu-Wakamatsu, Fukushima, Japan
| | - Satoshi HOSODA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Yu-ichi IIJIMA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Hitoshi IKEDA
- Research and Development Directorate, JAXA, Sagamihara, Kanagawa, Japan
| | - Masateru ISHIGURO
- Department of Physics and Astronomy, Seoul National University, Seoul, Korea
| | - Yoshiaki ISHIHARA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Takahiro IWATA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- The Graduate University for Advanced Studies (SOKENDAI), Hayama, Kanagawa, Japan
| | - Kosuke KAWAHARA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Shota KIKUCHI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- Planetary Exploration Research Center (PERC), Chiba Institute of Technology, Narashino, Chiba, Japan
| | - Kohei KITAZATO
- Faculty of Computer Science and Engineering, The University of Aizu, Aizu-Wakamatsu, Fukushima, Japan
| | - Koji MATSUMOTO
- National Astronomical Observatory of Japan, Mitaka, Tokyo, Japan
| | - Moe MATSUOKA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- Observatoire de Paris, Meudon, France
| | - Tatsuhiro MICHIKAMI
- Faculty of Engineering, Kindai University, Higashi-Hiroshima, Hiroshima, Japan
| | - Yuya MIMASU
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Akira MIURA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Tomokatsu MOROTA
- Graduate School of Environmental Studies, Nagoya University, Nagoya, Aichi, Japan
| | - Satoru NAKAZAWA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Noriyuki NAMIKI
- National Astronomical Observatory of Japan, Mitaka, Tokyo, Japan
| | - Hirotomo NODA
- National Astronomical Observatory of Japan, Mitaka, Tokyo, Japan
| | - Rina NOGUCHI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- Faculty of Science, Niigata University, Niigata, Japan
| | - Naoko OGAWA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- JAXA Space Exploration Center, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Kazunori OGAWA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Chisato OKAMOTO
- Graduate School of Science, Kobe University, Kobe, Hyogo, Japan
| | - Go ONO
- Research and Development Directorate, JAXA, Sagamihara, Kanagawa, Japan
| | - Masanobu OZAKI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Takanao SAIKI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | | | - Hirotaka SAWADA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Hiroki SENSHU
- Planetary Exploration Research Center (PERC), Chiba Institute of Technology, Narashino, Chiba, Japan
| | - Yuri SHIMAKI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Kei SHIRAI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- Graduate School of Science, Kobe University, Kobe, Hyogo, Japan
| | - Yuto TAKEI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Hiroshi TAKEUCHI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Satoshi TANAKA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- The Graduate University for Advanced Studies (SOKENDAI), Hayama, Kanagawa, Japan
- The University of Tokyo, Kashiwa, Chiba, Japan
| | - Eri TATSUMI
- Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Instituto de Astrofisica de Canarias, University of La Laguna, Tenerife, Spain
| | - Fuyuto TERUI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- Faculty of Engineering, Kanagawa Institute of Technology, Atsugi, Kanagawa, Japan
| | - Ryudo TSUKIZAKI
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Koji WADA
- Planetary Exploration Research Center (PERC), Chiba Institute of Technology, Narashino, Chiba, Japan
| | - Manabu YAMADA
- Planetary Exploration Research Center (PERC), Chiba Institute of Technology, Narashino, Chiba, Japan
| | - Tetsuya YAMADA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Yukio YAMAMOTO
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Hajime YANO
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Yasuhiro YOKOTA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Keisuke YOSHIHARA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Makoto YOSHIKAWA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- The Graduate University for Advanced Studies (SOKENDAI), Hayama, Kanagawa, Japan
| | - Kent YOSHIKAWA
- Research and Development Directorate, JAXA, Sagamihara, Kanagawa, Japan
| | - Masaki FUJIMOTO
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
| | - Sei-ichiro WATANABE
- Graduate School of Environmental Studies, Nagoya University, Nagoya, Aichi, Japan
| | - Yuichi TSUDA
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Sagamihara, Kanagawa, Japan
- Graduate School of Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
46
|
Chen Y, Loredo A, Chung A, Zhang M, Liu R, Xiao H. Biosynthesis and Genetic Incorporation of 3,4-Dihydroxy-L-Phenylalanine into Proteins in Escherichia coli. J Mol Biol 2021; 434:167412. [PMID: 34942167 PMCID: PMC9018569 DOI: 10.1016/j.jmb.2021.167412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/28/2022]
Abstract
While 20 canonical amino acids are used by most organisms for protein synthesis, the creation of cells that can use noncanonical amino acids (ncAAs) as additional protein building blocks holds great promise for preparing novel medicines and for studying complex questions in biological systems. However, only a small number of biosynthetic pathways for ncAAs have been reported to date, greatly restricting our ability to generate cells with ncAA building blocks. In this study, we report the creation of a completely autonomous bacterium that utilizes 3,4-dihydroxy-L-phenylalanine (DOPA) as its 21st amino acid building block. Like canonical amino acids, DOPA can be biosynthesized without exogenous addition and can be genetically incorporated into proteins in a site-specific manner. Equally important, the protein production yield of DOPA-containing proteins from these autonomous cells is greater than that of cells exogenously fed with 9 mM DOPA. The unique catechol moiety of DOPA can be used as a versatile handle for site-specific protein functionalizations via either oxidative coupling or strain-promoted oxidation-controlled cyclooctyne-1,2-quinone (SPOCQ) cycloaddition reactions. We further demonstrate the use of these autonomous cells in preparing fluorophore-labeled anti-human epidermal growth factor 2 (HER2) antibodies for the detection of HER2 expression on cancer cells.
Collapse
Affiliation(s)
- Yuda Chen
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005
| | - Axel Loredo
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005
| | - Anna Chung
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005
| | - Mengxi Zhang
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005
| | - Rui Liu
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005
| | - Han Xiao
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas, 77005; Department of Biosciences, Rice University, 6100 Main Street, Houston, Texas, 77005; Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005.
| |
Collapse
|
47
|
Woldegiorgis AG, Han Z, Lin X. Chiral Phosphoric Acid‐Catalyzed Enantioselective Synthesis of Pyrazole‐Based Unnatural α‐Amino Acid Derivatives. Adv Synth Catal 2021. [DOI: 10.1002/adsc.202101011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Alemayehu Gashaw Woldegiorgis
- Center of Chemistry for Frontier Technologies, Department of Chemistry Zhejiang University Hangzhou 310027 People's Republic of China
| | - Zhao Han
- Center of Chemistry for Frontier Technologies, Department of Chemistry Zhejiang University Hangzhou 310027 People's Republic of China
| | - Xufeng Lin
- Center of Chemistry for Frontier Technologies, Department of Chemistry Zhejiang University Hangzhou 310027 People's Republic of China
| |
Collapse
|
48
|
Hemkemeyer M, Schwalb SA, Heinze S, Joergensen RG, Wichern F. Functions of elements in soil microorganisms. Microbiol Res 2021; 252:126832. [PMID: 34508963 DOI: 10.1016/j.micres.2021.126832] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 12/15/2022]
Abstract
The soil microbial community fulfils various functions, such as nutrient cycling and carbon (C) sequestration, therefore contributing to maintenance of soil fertility and mitigation of global warming. In this context, a major focus of research has been on C, nitrogen (N) and phosphorus (P) cycling. However, from aquatic and other environments, it is well known that other elements beyond C, N, and P are essential for microbial functioning. Nonetheless, for soil microorganisms this knowledge has not yet been synthesised. To gain a better mechanistic understanding of microbial processes in soil systems, we aimed at summarising the current knowledge on the function of a range of essential or beneficial elements, which may affect the efficiency of microbial processes in soil. This knowledge is discussed in the context of microbial driven nutrient and C cycling. Our findings may support future investigations and data evaluation, where other elements than C, N, and P affect microbial processes.
Collapse
Affiliation(s)
- Michael Hemkemeyer
- Department of Soil Science and Plant Nutrition, Institute of Biogenic Resources in Sustainable Food Systems - From Farm to Function, Rhine-Waal University of Applied Sciences, Marie-Curie-Str. 1, 47533 Kleve, Germany.
| | - Sanja A Schwalb
- Department of Soil Science and Plant Nutrition, Institute of Biogenic Resources in Sustainable Food Systems - From Farm to Function, Rhine-Waal University of Applied Sciences, Marie-Curie-Str. 1, 47533 Kleve, Germany
| | - Stefanie Heinze
- Department of Soil Science & Soil Ecology, Ruhr-University Bochum, Universitätsstr. 150, 44801 Bochum, Germany
| | - Rainer Georg Joergensen
- Department of Soil Biology and Plant Nutrition, University of Kassel, Nordbahnhofstr. 1a, 37213 Witzenhausen, Germany
| | - Florian Wichern
- Department of Soil Science and Plant Nutrition, Institute of Biogenic Resources in Sustainable Food Systems - From Farm to Function, Rhine-Waal University of Applied Sciences, Marie-Curie-Str. 1, 47533 Kleve, Germany
| |
Collapse
|
49
|
Chandrashekar HB, Dolui P, Li B, Mandal A, Liu H, Guin S, Ge H, Maiti D. Ligand‐Enabled δ‐C(sp
3
)−H Borylation of Aliphatic Amines. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202105204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
| | - Pravas Dolui
- Department of Chemistry Indian Institute of Technology Bombay, Powai Mumbai 400076 India
| | - Bijin Li
- Department of Chemistry and Biochemistry Lubbock TX 79409-1061 USA
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Astam Mandal
- Department of Chemistry Indian Institute of Technology Bombay, Powai Mumbai 400076 India
| | - Hao Liu
- Department of Chemistry and Biochemistry Lubbock TX 79409-1061 USA
| | - Srimanta Guin
- Department of Chemistry Indian Institute of Technology Bombay, Powai Mumbai 400076 India
| | - Haibo Ge
- Department of Chemistry and Biochemistry Lubbock TX 79409-1061 USA
| | - Debabrata Maiti
- Department of Chemistry Indian Institute of Technology Bombay, Powai Mumbai 400076 India
| |
Collapse
|
50
|
Chandrashekar HB, Dolui P, Li B, Mandal A, Liu H, Guin S, Ge H, Maiti D. Ligand-Enabled δ-C(sp 3 )-H Borylation of Aliphatic Amines. Angew Chem Int Ed Engl 2021; 60:18194-18200. [PMID: 34117691 DOI: 10.1002/anie.202105204] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/18/2021] [Indexed: 02/03/2023]
Abstract
Directed C-H functionalization has been realized as a complimentary technique to achieve borylation at a distal position of aliphatic amines. Here, we demonstrated the oxidative borylation at the distal δ-position of aliphatic amines using various borylating agents, a palladium catalyst, and a rightly tuned ligand in the presence of a cheap oxidant. Moreover, an organopalladium δ-C(sp3 )-H-activated intermediate has been isolated and crystallographically characterized to get mechanistic insight.
Collapse
Affiliation(s)
| | - Pravas Dolui
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
| | - Bijin Li
- Department of Chemistry and Biochemistry, Lubbock, TX, 79409-1061, USA.,Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China
| | - Astam Mandal
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
| | - Hao Liu
- Department of Chemistry and Biochemistry, Lubbock, TX, 79409-1061, USA
| | - Srimanta Guin
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
| | - Haibo Ge
- Department of Chemistry and Biochemistry, Lubbock, TX, 79409-1061, USA
| | - Debabrata Maiti
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
| |
Collapse
|