1
|
Reinke PYA, Heiringhoff RS, Reindl T, Baker K, Taft MH, Meents A, Mulvihill DP, Davies OR, Fedorov R, Zahn M, Manstein DJ. Crystal structures of cables formed by the acetylated and unacetylated forms of the Schizosaccharomyces pombe tropomyosin ortholog Tpm Cdc8. J Biol Chem 2024; 300:107925. [PMID: 39461476 PMCID: PMC11626781 DOI: 10.1016/j.jbc.2024.107925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/09/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024] Open
Abstract
Cables formed by head-to-tail polymerization of tropomyosin, localized along the length of sarcomeric and cytoskeletal actin filaments, play a key role in regulating a wide range of motile and contractile processes. The stability of tropomyosin cables, their interaction with actin filaments and the functional properties of the resulting co-filaments are thought to be affected by N-terminal acetylation of tropomyosin. Here, we present high-resolution structures of cables formed by acetylated and unacetylated Schizosaccharomyces pombe tropomyosin ortholog TpmCdc8. The crystal structures represent different types of cables, each consisting of TpmCdc8 homodimers in a different conformation. The structures show how the interactions of the residues in the overlap junction contribute to cable formation and how local structural perturbations affect the conformational dynamics of the protein and its ability to transmit allosteric signals. In particular, N-terminal acetylation increases the helicity of the adjacent region, which leads to a local reduction in conformational dynamics and consequently to less fraying of the N-terminal region. This creates a more consistent complementary surface facilitating the formation of specific interactions across the overlap junction.
Collapse
Affiliation(s)
- Patrick Y A Reinke
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany; Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany; FS-BMX, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Robin S Heiringhoff
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany; Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
| | - Theresia Reindl
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany
| | - Karen Baker
- School of Biosciences, University of Kent, Canterbury, Kent, UK
| | - Manuel H Taft
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany
| | - Alke Meents
- FS-BMX, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | | | - Owen R Davies
- Wellcome Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh, UK
| | - Roman Fedorov
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany; Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
| | - Michael Zahn
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany; Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
| | - Dietmar J Manstein
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany; Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
2
|
Gagat P, Ostrówka M, Duda-Madej A, Mackiewicz P. Enhancing Antimicrobial Peptide Activity through Modifications of Charge, Hydrophobicity, and Structure. Int J Mol Sci 2024; 25:10821. [PMID: 39409150 PMCID: PMC11476776 DOI: 10.3390/ijms251910821] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Antimicrobial peptides (AMPs) are emerging as a promising alternative to traditional antibiotics due to their ability to disturb bacterial membranes and/or their intracellular processes, offering a potential solution to the growing problem of antimicrobial resistance. AMP effectiveness is governed by factors such as net charge, hydrophobicity, and the ability to form amphipathic secondary structures. When properly balanced, these characteristics enable AMPs to selectively target bacterial membranes while sparing eukaryotic cells. This review focuses on the roles of positive charge, hydrophobicity, and structure in influencing AMP activity and toxicity, and explores strategies to optimize them for enhanced therapeutic potential. We highlight the delicate balance between these properties and how various modifications, including amino acid substitutions, peptide tagging, or lipid conjugation, can either enhance or impair AMP performance. Notably, an increase in these parameters does not always yield the best results; sometimes, a slight reduction in charge, hydrophobicity, or structural stability improves the overall AMP therapeutic potential. Understanding these complex interactions is key to developing AMPs with greater antimicrobial activity and reduced toxicity, making them viable candidates in the fight against antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Przemysław Gagat
- Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-137 Wroclaw, Poland; (M.O.); (P.M.)
| | - Michał Ostrówka
- Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-137 Wroclaw, Poland; (M.O.); (P.M.)
| | - Anna Duda-Madej
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, Chalubinskiego 4, 50-368 Wroclaw, Poland;
| | - Paweł Mackiewicz
- Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-137 Wroclaw, Poland; (M.O.); (P.M.)
| |
Collapse
|
3
|
Nikolsky KS, Kulikova LI, Petrovskiy DV, Rudnev VR, Butkova TV, Malsagova KA, Kopylov AT, Kaysheva AL. Three-helix bundle and SH3-type barrels: autonomously stable structural motifs in small and large proteins. J Biomol Struct Dyn 2024; 42:9090-9104. [PMID: 37640007 DOI: 10.1080/07391102.2023.2250450] [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: 05/26/2023] [Accepted: 08/12/2023] [Indexed: 08/31/2023]
Abstract
In this study, we investigated two variants of a three-helix bundle and SH3-type barrel, compact in space, present in small and large proteins of various living organisms. Using a neural graph network, proteins with three-helix bundle (n = 1377) and SH3-type barrels (n = 1914) spatial folds were selected. Molecular experiments were performed for small proteins with these folds, and motifs were studied autonomously outside the protein environment at 300, 340, and 370 K. A comparative analysis of the main parameters of the structures in the course of the experiment was performed, including gyration radius, area accessible to the solvent, number of hydrophobic and hydrogen bonds, and root-mean-square deviation of atomic positions (RMSD). We exhibited an autonomous stability of the studied folds outside the protein environment in an aquatic medium. We aimed to demonstrate the possibility of analyzing three-helix bundle and SH3-type barrels autonomously outside the protein globule, thereby reducing the computational time and increasing performance without significant loss of information.Communicated by Ramaswamy H. Sarma.
Collapse
|
4
|
Gray V, Letteri RA. Designing Coiled Coils for Heterochiral Complexation to Enhance Binding and Enzymatic Stability. Biomacromolecules 2024; 25:5273-5280. [PMID: 38980285 PMCID: PMC11323006 DOI: 10.1021/acs.biomac.4c00661] [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: 05/17/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
Coiled coils, commonly found in native proteins, are helical motifs important for mediating intermolecular interactions. While coiled coils are attractive for use in new therapies and biomaterials, the lack of enzymatic stability of naturally occurring l-peptides may limit their implementation in biological environments. d-peptides are of interest for biomedical applications as they are resistant to enzymatic degradation and recent reports indicate that stereochemistry-driven interactions, achieved by blending d- and l-peptides, yield access to a greater range of binding affinities and a resistance to enzymatic degradation compared to l-peptides alone. To our knowledge, this effect has not been studied in coiled coils. Here, we investigate the effects of blending heterochiral E/K coiled coils, which are a set of coiled coils widely used in biomaterials. We found that we needed to redesign the coiled coils from a repeating pattern of seven amino acids (heptad) to a repeating pattern of 11 amino acids (hendecad) to make them more amenable to heterochiral complex formation. The redesigned hendecad coiled coils form both homochiral and heterochiral complexes, where the heterochiral complexes have stronger heats of binding between the constituent peptides and are more enzymatically stable than the analogous homochiral complexes. Our results highlight the ability to design peptides to make them amenable to heterochiral complexation, so as to achieve desirable properties like increased enzymatic stability and stronger binding. Looking forward, understanding how to engineer peptides to utilize stereochemistry as a materials design tool will be important to the development of next-generation therapeutics and biomaterials.
Collapse
Affiliation(s)
- Vincent
P. Gray
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Rachel A. Letteri
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903, United States
| |
Collapse
|
5
|
Chinthalapudi K, Heissler SM. Structure, regulation, and mechanisms of nonmuscle myosin-2. Cell Mol Life Sci 2024; 81:263. [PMID: 38878079 PMCID: PMC11335295 DOI: 10.1007/s00018-024-05264-6] [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: 03/11/2024] [Revised: 04/24/2024] [Accepted: 04/30/2024] [Indexed: 06/23/2024]
Abstract
Members of the myosin superfamily of molecular motors are large mechanochemical ATPases that are implicated in an ever-expanding array of cellular functions. This review focuses on mammalian nonmuscle myosin-2 (NM2) paralogs, ubiquitous members of the myosin-2 family of filament-forming motors. Through the conversion of chemical energy into mechanical work, NM2 paralogs remodel and shape cells and tissues. This process is tightly controlled in time and space by numerous synergetic regulation mechanisms to meet cellular demands. We review how recent advances in structural biology together with elegant biophysical and cell biological approaches have contributed to our understanding of the shared and unique mechanisms of NM2 paralogs as they relate to their kinetics, regulation, assembly, and cellular function.
Collapse
Affiliation(s)
- Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Sarah M Heissler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH, 43210, USA.
| |
Collapse
|
6
|
Yang T, Wang Y, Jiang J, Wang P, Zhong Y, Zhou Q, Wang X, Cai J, Huang M, Jiang D, Dai T, Cao W. Influence of High-Molecular-Weight Glutenin Subunit on Components and Multiscale Structure of Gluten and Dough Quality in Soft Wheat. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:4943-4956. [PMID: 36924464 DOI: 10.1021/acs.jafc.2c07958] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
A set of high-molecular-weight glutenin subunit (HMW-GS) deletion lines were used to investigate the influences of HMW-GS on wheat gluten, and dough properties were investigated using a set of HMW-GS deletion lines. Results showed that HMW-GS deletion significantly decreased the dough stability time, as well as viscoelastic moduli (G' and G″), compared with the wild type, where the deletion of x-type HMW-GSs (Ax1d, Bx7d, and Dy12d) decreased more than y-type HMW-GSs (By8d and Dy12d). The deletion of HMW-GS significantly decreased HMW-GS contents and increased α-/γ-gliadin contents. A proteomic study showed that the HMW-GS deletion down-regulated the HMW-GS, β-amylase, serpins, and protein disulfide isomerase and up-regulated the LMW-GS, α/γ-gliadin, and α-amylase inhibitor. Meanwhile, HMW-GS deletion significantly decreased contents of β-turn and β-sheet. In addition, less energetically stable disulfide conformations (trans-gauche-gauche and trans-gauche-trans) were abundant in HMW-GS deletion lines. Furthermore, analysis of five HMW-GSs based on amino acid sequences proved that Dx2 and Bx7 had a more stable structure, followed by Ax1, then Dy12, and finally By8.
Collapse
Affiliation(s)
- Tao Yang
- National Technique Innovation Center for Regional Wheat Production/Key Laboratory of Crop Physiology, Ecology and Management, Ministry of Agriculture/National Engineering and Technology Center for Information Agriculture, College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Yingpeng Wang
- National Technique Innovation Center for Regional Wheat Production/Key Laboratory of Crop Physiology, Ecology and Management, Ministry of Agriculture/National Engineering and Technology Center for Information Agriculture, College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Jiali Jiang
- National Technique Innovation Center for Regional Wheat Production/Key Laboratory of Crop Physiology, Ecology and Management, Ministry of Agriculture/National Engineering and Technology Center for Information Agriculture, College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Pei Wang
- National Technique Innovation Center for Regional Wheat Production/Key Laboratory of Crop Physiology, Ecology and Management, Ministry of Agriculture/National Engineering and Technology Center for Information Agriculture, College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Yingxin Zhong
- National Technique Innovation Center for Regional Wheat Production/Key Laboratory of Crop Physiology, Ecology and Management, Ministry of Agriculture/National Engineering and Technology Center for Information Agriculture, College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Qin Zhou
- National Technique Innovation Center for Regional Wheat Production/Key Laboratory of Crop Physiology, Ecology and Management, Ministry of Agriculture/National Engineering and Technology Center for Information Agriculture, College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Xiao Wang
- National Technique Innovation Center for Regional Wheat Production/Key Laboratory of Crop Physiology, Ecology and Management, Ministry of Agriculture/National Engineering and Technology Center for Information Agriculture, College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Jian Cai
- National Technique Innovation Center for Regional Wheat Production/Key Laboratory of Crop Physiology, Ecology and Management, Ministry of Agriculture/National Engineering and Technology Center for Information Agriculture, College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Mei Huang
- National Technique Innovation Center for Regional Wheat Production/Key Laboratory of Crop Physiology, Ecology and Management, Ministry of Agriculture/National Engineering and Technology Center for Information Agriculture, College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Dong Jiang
- National Technique Innovation Center for Regional Wheat Production/Key Laboratory of Crop Physiology, Ecology and Management, Ministry of Agriculture/National Engineering and Technology Center for Information Agriculture, College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Tingbo Dai
- National Technique Innovation Center for Regional Wheat Production/Key Laboratory of Crop Physiology, Ecology and Management, Ministry of Agriculture/National Engineering and Technology Center for Information Agriculture, College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Weixing Cao
- National Technique Innovation Center for Regional Wheat Production/Key Laboratory of Crop Physiology, Ecology and Management, Ministry of Agriculture/National Engineering and Technology Center for Information Agriculture, College of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| |
Collapse
|
7
|
Risi CM, Belknap B, White HD, Dryden K, Pinto JR, Chase PB, Galkin VE. High-resolution cryo-EM structure of the junction region of the native cardiac thin filament in relaxed state. PNAS NEXUS 2023; 2:pgac298. [PMID: 36712934 PMCID: PMC9832952 DOI: 10.1093/pnasnexus/pgac298] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Cardiac contraction depends on molecular interactions among sarcomeric proteins coordinated by the rising and falling intracellular Ca2+ levels. Cardiac thin filament (cTF) consists of two strands composed of actin, tropomyosin (Tm), and equally spaced troponin (Tn) complexes forming regulatory units. Tn binds Ca2+ to move Tm strand away from myosin-binding sites on actin to enable actomyosin cross-bridges required for force generation. The Tn complex has three subunits-Ca2+-binding TnC, inhibitory TnI, and Tm-binding TnT. Tm strand is comprised of adjacent Tm molecules that overlap "head-to-tail" along the actin filament. The N-terminus of TnT (e.g., TnT1) binds to the Tm overlap region to form the cTF junction region-the region that connects adjacent regulatory units and confers to cTF internal cooperativity. Numerous studies have predicted interactions among actin, Tm, and TnT1 within the junction region, although a direct structural description of the cTF junction region awaited completion. Here, we report a 3.8 Å resolution cryo-EM structure of the native cTF junction region at relaxing (pCa 8) Ca2+ conditions. We provide novel insights into the "head-to-tail" interactions between adjacent Tm molecules and interactions between the Tm junction with F-actin. We demonstrate how TnT1 stabilizes the Tm overlap region via its interactions with the Tm C- and N-termini and actin. Our data show that TnT1 works as a joint that anchors the Tm overlap region to actin, which stabilizes the relaxed state of the cTF. Our structure provides insight into the molecular basis of cardiac diseases caused by missense mutations in TnT1.
Collapse
Affiliation(s)
- Cristina M Risi
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Betty Belknap
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Howard D White
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Kelly Dryden
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22904, USA
| | - Jose R Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32304, USA
| | - P Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Vitold E Galkin
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| |
Collapse
|
8
|
Park OS, Bang JK, Cheong C, Jeon YH. Structure of AQEE-30 of VGF Neuropeptide in Membrane-Mimicking Environments. Int J Mol Sci 2022; 23:ijms232213953. [PMID: 36430431 PMCID: PMC9696787 DOI: 10.3390/ijms232213953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
AQEE-30 is one of the VGF peptides, which are derived from the VGF polypeptide precursor, and related to various physiological phenomena including neuroprotective effects in Huntington's disease and amyotrophic lateral sclerosis (ALS). Although various functions of AQEE-30 have been reported so far, the structure of this peptide has not been reported yet. In this study, the structure of human AQEE-30 was investigated in hexafluoroisopropanol (HFIP) and dodecyl phosphocholine (DPC) micelle solutions, using circular dichroism (CD) and nuclear magnetic resonance (NMR) spectroscopy. CD results showed that AQEE-30 had a partial helical structure in aqueous buffer, and the helical structure was stabilized in the HFIP and DPC micelle solutions. The 3D structures determined by NMR spectroscopy showed that AQEE-30 adopted mainly α-helical structure in both the HFIP and DPC micelle solutions. The surface of AQEE-30 showed that it was predominantly negatively charged. The residues from 601 to 611 in both the HFIP and DPC micelle solutions showed amphiphilicity with four negatively charged residues, glutamate. The C-terminal consecutive arginine residues formed a partial positively charged surface. These results suggest an α-helical active structure of AQEE-30 in the cell-membrane environment.
Collapse
Affiliation(s)
- One-Sung Park
- College of Pharmacy, Korea University Sejong Campus, Sejong 30019, Korea
- Division of Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Korea
| | - Jeong-Kyu Bang
- Division of Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Korea
| | - Chaejoon Cheong
- Division of Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Korea
- Department of Bioanalytical Sciences, University of Science and Technology, Cheongju 28119, Korea
- Correspondence: (C.C.); (Y.-H.J.)
| | - Young-Ho Jeon
- College of Pharmacy, Korea University Sejong Campus, Sejong 30019, Korea
- Correspondence: (C.C.); (Y.-H.J.)
| |
Collapse
|
9
|
Gopinathan Nair A, Rabas N, Lejon S, Homiski C, Osborne MJ, Cyr N, Sverzhinsky A, Melendy T, Pascal JM, Laue ED, Borden KLB, Omichinski JG, Verreault A. Unorthodox PCNA Binding by Chromatin Assembly Factor 1. Int J Mol Sci 2022; 23:11099. [PMID: 36232396 PMCID: PMC9570017 DOI: 10.3390/ijms231911099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Abstract
The eukaryotic DNA replication fork is a hub of enzymes that continuously act to synthesize DNA, propagate DNA methylation and other epigenetic marks, perform quality control, repair nascent DNA, and package this DNA into chromatin. Many of the enzymes involved in these spatiotemporally correlated processes perform their functions by binding to proliferating cell nuclear antigen (PCNA). A long-standing question has been how the plethora of PCNA-binding enzymes exert their activities without interfering with each other. As a first step towards deciphering this complex regulation, we studied how Chromatin Assembly Factor 1 (CAF-1) binds to PCNA. We demonstrate that CAF-1 binds to PCNA in a heretofore uncharacterized manner that depends upon a cation-pi (π) interaction. An arginine residue, conserved among CAF-1 homologs but absent from other PCNA-binding proteins, inserts into the hydrophobic pocket normally occupied by proteins that contain canonical PCNA interaction peptides (PIPs). Mutation of this arginine disrupts the ability of CAF-1 to bind PCNA and to assemble chromatin. The PIP of the CAF-1 p150 subunit resides at the extreme C-terminus of an apparent long α-helix (119 amino acids) that has been reported to bind DNA. The length of that helix and the presence of a PIP at the C-terminus are evolutionarily conserved among numerous species, ranging from yeast to humans. This arrangement of a very long DNA-binding coiled-coil that terminates in PIPs may serve to coordinate DNA and PCNA binding by CAF-1.
Collapse
Affiliation(s)
- Amogh Gopinathan Nair
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC H3T 1J4, Canada
- Molecular Biology Program, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Nick Rabas
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK
| | - Sara Lejon
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK
| | - Caleb Homiski
- Departments of Biochemistry and Microbiology & Immunology, University at Buffalo Jacobs School of Medicine & Biomedical Sciences, 955 Main Street, Buffalo, NY 14210, USA
| | - Michael J. Osborne
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Normand Cyr
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Aleksandr Sverzhinsky
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Thomas Melendy
- Departments of Biochemistry and Microbiology & Immunology, University at Buffalo Jacobs School of Medicine & Biomedical Sciences, 955 Main Street, Buffalo, NY 14210, USA
| | - John M. Pascal
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Ernest D. Laue
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK
| | - Katherine L. B. Borden
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC H3T 1J4, Canada
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - James G. Omichinski
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Alain Verreault
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC H3T 1J4, Canada
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
10
|
Ayinuola O, Ayinuola YA, Qiu C, Lee SW, Ploplis VA, Castellino FJ. Binding of the kringle-2 domain of human plasminogen to streptococcal PAM-type M-protein causes dissociation of PAM dimers. Microbiologyopen 2021; 10:e1252. [PMID: 34964287 PMCID: PMC8633249 DOI: 10.1002/mbo3.1252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/03/2021] [Indexed: 11/24/2022] Open
Abstract
The direct binding of human plasminogen (hPg), via its kringle-2 domain (K2hPg ), to streptococcal M-protein (PAM), largely contributes to the pathogenesis of Pattern D Group A Streptococcus pyogenes (GAS). However, the mechanism of complex formation is unknown. In a system consisting of a Class II PAM from Pattern D GAS isolate NS88.2 (PAMNS88.2 ), with one K2hPg binding a-repeat in its A-domain, we employed biophysical techniques to analyze the mechanism of the K2hPg /PAMNS88.2 interaction. We show that apo-PAMNS88.2 is a coiled-coil homodimer (M.Wt. ~80 kDa) at 4°C-25°C, and is monomeric (M.Wt. ~40 kDa) at 37°C, demonstrating a temperature-dependent dissociation of PAMNS88.2 over a narrow temperature range. PAMNS88.2 displayed a single tight binding site for K2hPg at 4°C, which progressively increased at 25°C through 37°C. We isolated the K2hPg /PAMNS88.2 complexes at 4°C, 25°C, and 37°C and found molecular weights of ~50 kDa at each temperature, corresponding to a 1:1 (m:m) K2hPg /PAMNS88.2 monomer complex. hPg activation experiments by streptokinase demonstrated that the hPg/PAMNS88.2 monomer complexes are fully functional. The data show that PAM dimers dissociate into functional monomers at physiological temperatures or when presented with the active hPg module (K2hPg ) showing that PAM is a functional monomer at 37°C.
Collapse
Affiliation(s)
- Olawole Ayinuola
- W. M. Keck Center for Transgene ResearchUniversity of Notre DameNotre DameIndianaUSA
| | - Yetunde A. Ayinuola
- W. M. Keck Center for Transgene ResearchUniversity of Notre DameNotre DameIndianaUSA
| | - Cunjia Qiu
- W. M. Keck Center for Transgene ResearchUniversity of Notre DameNotre DameIndianaUSA
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Shaun W. Lee
- W. M. Keck Center for Transgene ResearchUniversity of Notre DameNotre DameIndianaUSA
- Department of Biological SciencesUniversity of Notre DameNotre DameIndianaUSA
| | - Victoria A. Ploplis
- W. M. Keck Center for Transgene ResearchUniversity of Notre DameNotre DameIndianaUSA
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Francis J. Castellino
- W. M. Keck Center for Transgene ResearchUniversity of Notre DameNotre DameIndianaUSA
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| |
Collapse
|
11
|
Ultsch M, Holliday MJ, Gerhardy S, Moran P, Scales SJ, Gupta N, Oltrabella F, Chiu C, Fairbrother W, Eigenbrot C, Kirchhofer D. Structures of the ApoL1 and ApoL2 N-terminal domains reveal a non-classical four-helix bundle motif. Commun Biol 2021; 4:916. [PMID: 34316015 PMCID: PMC8316464 DOI: 10.1038/s42003-021-02387-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Apolipoprotein L1 (ApoL1) is a circulating innate immunity protein protecting against trypanosome infection. However, two ApoL1 coding variants are associated with a highly increased risk of chronic kidney disease. Here we present X-ray and NMR structures of the N-terminal domain (NTD) of ApoL1 and of its closest relative ApoL2. In both proteins, four of the five NTD helices form a four-helix core structure which is different from the classical four-helix bundle and from the pore-forming domain of colicin A. The reactivity with a conformation-specific antibody and structural models predict that this four-helix motif is also present in the NTDs of ApoL3 and ApoL4, suggesting related functions within the small ApoL family. The long helix 5 of ApoL1 is conformationally flexible and contains the BH3-like region. This BH3-like α-helix resembles true BH3 domains only in sequence and structure but not in function, since it does not bind to the pro-survival members of the Bcl-2 family, suggesting a Bcl-2-independent role in cytotoxicity. These findings should expedite a more comprehensive structural and functional understanding of the ApoL immune protein family.
Collapse
Affiliation(s)
- Mark Ultsch
- Department of Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | - Michael J Holliday
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA
| | - Stefan Gerhardy
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA
| | - Paul Moran
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA
| | - Suzie J Scales
- Department of Immunology, Genentech Inc., South San Francisco, CA, USA
| | - Nidhi Gupta
- Department of Immunology, Genentech Inc., South San Francisco, CA, USA
| | | | - Cecilia Chiu
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Wayne Fairbrother
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA
| | - Charles Eigenbrot
- Department of Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | - Daniel Kirchhofer
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA.
| |
Collapse
|
12
|
Gupta K, Wen Y, Ninan NS, Raimer AC, Sharp R, Spring A, Sarachan KL, Johnson MC, Van Duyne GD, Matera AG. Assembly of higher-order SMN oligomers is essential for metazoan viability and requires an exposed structural motif present in the YG zipper dimer. Nucleic Acids Res 2021; 49:7644-7664. [PMID: 34181727 PMCID: PMC8287954 DOI: 10.1093/nar/gkab508] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/03/2021] [Indexed: 12/16/2022] Open
Abstract
Protein oligomerization is one mechanism by which homogenous solutions can separate into distinct liquid phases, enabling assembly of membraneless organelles. Survival Motor Neuron (SMN) is the eponymous component of a large macromolecular complex that chaperones biogenesis of eukaryotic ribonucleoproteins and localizes to distinct membraneless organelles in both the nucleus and cytoplasm. SMN forms the oligomeric core of this complex, and missense mutations within its YG box domain are known to cause Spinal Muscular Atrophy (SMA). The SMN YG box utilizes a unique variant of the glycine zipper motif to form dimers, but the mechanism of higher-order oligomerization remains unknown. Here, we use a combination of molecular genetic, phylogenetic, biophysical, biochemical and computational approaches to show that formation of higher-order SMN oligomers depends on a set of YG box residues that are not involved in dimerization. Mutation of key residues within this new structural motif restricts assembly of SMN to dimers and causes locomotor dysfunction and viability defects in animal models.
Collapse
Affiliation(s)
- Kushol Gupta
- Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19105-6059, USA
| | - Ying Wen
- Integrative Program for Biological & Genome Sciences, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nisha S Ninan
- Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19105-6059, USA
| | - Amanda C Raimer
- Integrative Program for Biological & Genome Sciences, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Curriculum in Genetics and Molecular Biology, Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Robert Sharp
- Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19105-6059, USA
| | - Ashlyn M Spring
- Integrative Program for Biological & Genome Sciences, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kathryn L Sarachan
- Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19105-6059, USA
| | - Meghan C Johnson
- Department of Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Gregory D Van Duyne
- Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19105-6059, USA
| | - A Gregory Matera
- Integrative Program for Biological & Genome Sciences, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Curriculum in Genetics and Molecular Biology, Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
13
|
Rodriguez-Cabello JC, Gonzalez De Torre I, González-Pérez M, González-Pérez F, Montequi I. Fibrous Scaffolds From Elastin-Based Materials. Front Bioeng Biotechnol 2021; 9:652384. [PMID: 34336798 PMCID: PMC8323661 DOI: 10.3389/fbioe.2021.652384] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 06/25/2021] [Indexed: 11/28/2022] Open
Abstract
Current cutting-edge strategies in biomaterials science are focused on mimicking the design of natural systems which, over millions of years, have evolved to exhibit extraordinary properties. Based on this premise, one of the most challenging tasks is to imitate the natural extracellular matrix (ECM), due to its ubiquitous character and its crucial role in tissue integrity. The anisotropic fibrillar architecture of the ECM has been reported to have a significant influence on cell behaviour and function. A new paradigm that pivots around the idea of incorporating biomechanical and biomolecular cues into the design of biomaterials and systems for biomedical applications has emerged in recent years. Indeed, current trends in materials science address the development of innovative biomaterials that include the dynamics, biochemistry and structural features of the native ECM. In this context, one of the most actively studied biomaterials for tissue engineering and regenerative medicine applications are nanofiber-based scaffolds. Herein we provide a broad overview of the current status, challenges, manufacturing methods and applications of nanofibers based on elastin-based materials. Starting from an introduction to elastin as an inspiring fibrous protein, as well as to the natural and synthetic elastin-based biomaterials employed to meet the challenge of developing ECM-mimicking nanofibrous-based scaffolds, this review will follow with a description of the leading strategies currently employed in nanofibrous systems production, which in the case of elastin-based materials are mainly focused on supramolecular self-assembly mechanisms and the use of advanced manufacturing technologies. Thus, we will explore the tendency of elastin-based materials to form intrinsic fibers, and the self-assembly mechanisms involved. We will describe the function and self-assembly mechanisms of silk-like motifs, antimicrobial peptides and leucine zippers when incorporated into the backbone of the elastin-based biomaterial. Advanced polymer-processing technologies, such as electrospinning and additive manufacturing, as well as their specific features, will be presented and reviewed for the specific case of elastin-based nanofiber manufacture. Finally, we will present our perspectives and outlook on the current challenges facing the development of nanofibrous ECM-mimicking scaffolds based on elastin and elastin-like biomaterials, as well as future trends in nanofabrication and applications.
Collapse
Affiliation(s)
- Jose Carlos Rodriguez-Cabello
- BIOFORGE, University of Valladolid, Valladolid, Spain
- Center for Biomedical Research in the Network in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Israel Gonzalez De Torre
- BIOFORGE, University of Valladolid, Valladolid, Spain
- Center for Biomedical Research in the Network in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Miguel González-Pérez
- BIOFORGE, University of Valladolid, Valladolid, Spain
- Center for Biomedical Research in the Network in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Fernando González-Pérez
- BIOFORGE, University of Valladolid, Valladolid, Spain
- Center for Biomedical Research in the Network in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Irene Montequi
- BIOFORGE, University of Valladolid, Valladolid, Spain
- Center for Biomedical Research in the Network in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
14
|
Arnittali M, Rissanou AN, Amprazi M, Kokkinidis M, Harmandaris V. Structure and Thermal Stability of wtRop and RM6 Proteins through All-Atom Molecular Dynamics Simulations and Experiments. Int J Mol Sci 2021; 22:ijms22115931. [PMID: 34073028 PMCID: PMC8199364 DOI: 10.3390/ijms22115931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 01/07/2023] Open
Abstract
In the current work we study, via molecular simulations and experiments, the folding and stability of proteins from the tertiary motif of 4-α-helical bundles, a recurrent motif consisting of four amphipathic α-helices packed in a parallel or antiparallel fashion. The focus is on the role of the loop region in the structure and the properties of the wild-type Rop (wtRop) and RM6 proteins, exploring the key factors which can affect them, through all-atom molecular dynamics (MD) simulations and supporting by experimental findings. A detailed investigation of structural and conformational properties of wtRop and its RM6 loopless mutation is presented, which display different physical characteristics even in their native states. Then, the thermal stability of both proteins is explored showing RM6 as more thermostable than wtRop through all studied measures. Deviations from native structures are detected mostly in tails and loop regions and most flexible residues are indicated. Decrease of hydrogen bonds with the increase of temperature is observed, as well as reduction of hydrophobic contacts in both proteins. Experimental data from circular dichroism spectroscopy (CD), are also presented, highlighting the effect of temperature on the structural integrity of wtRop and RM6. The central goal of this study is to explore on the atomic level how a protein mutation can cause major changes in its physical properties, like its structural stability.
Collapse
Affiliation(s)
- Maria Arnittali
- Institute of Applied and Computational Mathematics (IACM), Foundation for Research and Technology Hellas (FORTH), IACM/FORTH, GR-71110 Heraklion, Crete, Greece; (M.A.); (V.H.)
- Department of Mathematics and Applied Mathematics, University of Crete, GR-71409 Heraklion, Crete, Greece
| | - Anastassia N. Rissanou
- Institute of Applied and Computational Mathematics (IACM), Foundation for Research and Technology Hellas (FORTH), IACM/FORTH, GR-71110 Heraklion, Crete, Greece; (M.A.); (V.H.)
- Department of Mathematics and Applied Mathematics, University of Crete, GR-71409 Heraklion, Crete, Greece
- Correspondence: ; Tel.: +30-2810-393746
| | - Maria Amprazi
- Department of Biology, University of Crete, GR-71409 Heraklion, Crete, Greece; (M.A.); (M.K.)
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, GR-70013 Heraklion, Crete, Greece
| | - Michael Kokkinidis
- Department of Biology, University of Crete, GR-71409 Heraklion, Crete, Greece; (M.A.); (M.K.)
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, GR-70013 Heraklion, Crete, Greece
| | - Vagelis Harmandaris
- Institute of Applied and Computational Mathematics (IACM), Foundation for Research and Technology Hellas (FORTH), IACM/FORTH, GR-71110 Heraklion, Crete, Greece; (M.A.); (V.H.)
- Department of Mathematics and Applied Mathematics, University of Crete, GR-71409 Heraklion, Crete, Greece
- Computation-Based Science and Technology Research Center, The Cyprus Institute, 2121 Nicosia, Cyprus
| |
Collapse
|
15
|
Orientational Ambiguity in Septin Coiled Coils and its Structural Basis. J Mol Biol 2021; 433:166889. [PMID: 33639214 DOI: 10.1016/j.jmb.2021.166889] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/25/2021] [Accepted: 02/17/2021] [Indexed: 12/21/2022]
Abstract
Septins are an example of subtle molecular recognition whereby different paralogues must correctly assemble into functional filaments important for essential cellular events such as cytokinesis. Most possess C-terminal domains capable of forming coiled coils which are believed to be involved in filament formation and bundling. Here, we report an integrated structural approach which aims to unravel their architectural diversity and in so doing provide direct structural information for the coiled-coil regions of five human septins. Unexpectedly, we encounter dimeric structures presenting both parallel and antiparallel arrangements which are in consonance with molecular modelling suggesting that both are energetically accessible. These sequences therefore code for two metastable states of different orientations which employ different but overlapping interfaces. The antiparallel structures present a mixed coiled-coil interface, one side of which is dominated by a continuous chain of core hydrophilic residues. This unusual type of coiled coil could be used to expand the toolkit currently available to the protein engineer for the design of previously unforeseen coiled-coil based assemblies. Within a physiological context, our data provide the first atomic details related to the assumption that the parallel orientation is likely formed between septin monomers from the same filament whilst antiparallelism may participate in the widely described interfilament cross bridges necessary for higher order structures and thereby septin function.
Collapse
|
16
|
Prediction of Protein-Protein Binding Interactions in Dimeric Coiled Coils by Information Contained in Folding Energy Landscapes. Int J Mol Sci 2021; 22:ijms22031368. [PMID: 33573048 PMCID: PMC7866404 DOI: 10.3390/ijms22031368] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 11/16/2022] Open
Abstract
Coiled coils represent the simplest form of a complex formed between two interacting protein partners. Their extensive study has led to the development of various methods aimed towards the investigation and design of complex forming interactions. Despite the progress that has been made to predict the binding affinities for protein complexes, and specifically those tailored towards coiled coils, many challenges still remain. In this work, we explore whether the information contained in dimeric coiled coil folding energy landscapes can be used to predict binding interactions. Using the published SYNZIP dataset, we start from the amino acid sequence, to simultaneously fold and dock approximately 1000 coiled coil dimers. Assessment of the folding energy landscapes showed that a model based on the calculated number of clusters for the lowest energy structures displayed a signal that correlates with the experimentally determined protein interactions. Although the revealed correlation is weak, we show that such correlation exists; however, more work remains to establish whether further improvements can be made to the presented model.
Collapse
|
17
|
Abstract
Fibrinogen is a large glycoprotein, synthesized primarily in the liver. With a normal plasma concentration of 1.5-3.5 g/L, fibrinogen is the most abundant blood coagulation factor. The final stage of blood clot formation is the conversion of soluble fibrinogen to insoluble fibrin, the polymeric scaffold for blood clots that stop bleeding (a protective reaction called hemostasis) or obstruct blood vessels (pathological thrombosis). Fibrin is a viscoelastic polymer and the structural and mechanical properties of the fibrin scaffold determine its effectiveness in hemostasis and the development and outcome of thrombotic complications. Fibrin polymerization comprises a number of consecutive reactions, each affecting the ultimate 3D porous network structure. The physical properties of fibrin clots are determined by structural features at the individual fibrin molecule, fibrin fiber, network, and whole clot levels and are among the most important functional characteristics, enabling the blood clot to withstand arterial blood flow, platelet-driven clot contraction, and other dynamic forces. This chapter describes the molecular structure of fibrinogen, the conversion of fibrinogen to fibrin, the mechanical properties of fibrin as well as its structural origins and lastly provides evidence for the role of altered fibrin clot properties in both thrombosis and bleeding.
Collapse
|
18
|
Nandakumar A, Ito Y, Ueda M. Solvent Effects on the Self-Assembly of an Amphiphilic Polypeptide Incorporating α-Helical Hydrophobic Blocks. J Am Chem Soc 2020; 142:20994-21003. [PMID: 33272014 DOI: 10.1021/jacs.0c03425] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The self-assembly of biological molecules is an important pathway to understanding the molecular basis of complex metabolic events. The presence of a cosolvent in an aqueous solution during the self-assembly process can promote the formation of kinetically trapped metastable intermediates. In nature, a category of cosolvents termed osmolytes can work to strengthen the hydrogen-bond network of water such that the native states of certain proteins are favored, thus modulating their function and stability. However, identifying cosolvents that act as osmolytes in biomimetic applications, such as the self-assembly of soft materials, remains challenging. The present work examined the effects of ethanol (EtOH) and acetonitrile (ACN) as cosolvents on the self-assembly of the amphiphilic polypeptide PSar30-(l-Leu-Aib)6 (S30L12), which incorporates α-helical hydrophobic blocks, in aqueous solution. The results provided a direct observation of morphological behavior of S30L12 as a function of solvent composition. Morphological transitions were investigated using transmission electron microscopy, while the packing of peptide molecules was assessed using circular dichroism analyses and evaluations of membrane fluidity. In the EtOH/H2O mixtures, the EtOH strengthened the hydrogen-bond network of the water, thus limiting the hydrophobic hydration of S30L12 assemblies and enhancing hydrophobic interactions between assemblies. In contrast, ACN formed self-associated nanoclusters in water and at the hydrophobic cores of peptide assemblies to stabilize the edges exposed to bulk water and enhance the assembly kinetics. Fourier transform infrared (FT-IR) analysis indicated that both EtOH and ACN can modify the self-assembly of biomaterials in the same manner as osmolyte protectants or denaturants.
Collapse
Affiliation(s)
- Avanashiappan Nandakumar
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science (CEMS), 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yoshihiro Ito
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science (CEMS), 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.,Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Motoki Ueda
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science (CEMS), 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.,Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
19
|
Pinter TBJ, Manickas EC, Tolbert AE, Koebke KJ, Deb A, Penner-Hahn JE, Pecoraro VL. Making or Breaking Metal-Dependent Catalytic Activity: The Role of Stammers in Designed Three-Stranded Coiled Coils. Angew Chem Int Ed Engl 2020; 59:20445-20449. [PMID: 32748510 PMCID: PMC7722090 DOI: 10.1002/anie.202008356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/21/2020] [Indexed: 12/31/2022]
Abstract
While many life-critical reactions would be infeasibly slow without metal cofactors, a detailed understanding of how protein structure can influence catalytic activity remains elusive. Using de novo designed three-stranded coiled coils (TRI and Grand peptides formed using a heptad repeat approach), we examine how the insertion of a three residue discontinuity, known as a stammer insert, directly adjacent to a (His)3 metal binding site alters catalytic activity. The stammer, which locally alters the twist of the helix, significantly increases copper-catalyzed nitrite reductase activity (CuNiR). In contrast, the well-established zinc-catalyzed carbonic anhydrase activity (p-nitrophenyl acetate, pNPA) is effectively ablated. This study illustrates how the perturbation of the protein sequence using non-coordinating and non-acid base residues in the helical core can perturb metalloenzyme activity through the simple expedient of modifying the helical pitch adjacent to the catalytic center.
Collapse
Affiliation(s)
- Tyler B. J. Pinter
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA, 48109
| | | | - Audrey E. Tolbert
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA, 48109
| | - Karl J. Koebke
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA, 48109
| | - Aniruddha Deb
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA, 48109
| | - James E. Penner-Hahn
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA, 48109
| | - Vincent L. Pecoraro
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA, 48109
| |
Collapse
|
20
|
Pinter TBJ, Manickas EC, Tolbert AE, Koebke KJ, Deb A, Penner‐Hahn JE, Pecoraro VL. Making or Breaking Metal‐Dependent Catalytic Activity: The Role of Stammers in Designed Three‐Stranded Coiled Coils. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202008356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | | | - Audrey E. Tolbert
- Department of Chemistry University of Michigan Ann Arbor MI 48109 USA
| | - Karl J. Koebke
- Department of Chemistry University of Michigan Ann Arbor MI 48109 USA
| | - Aniruddha Deb
- Department of Chemistry University of Michigan Ann Arbor MI 48109 USA
| | | | | |
Collapse
|
21
|
Li J, Zhao Y, Zhou P, Hu X, Wang D, King SM, Rogers SE, Wang J, Lu JR, Xu H. Ordered Nanofibers Fabricated from Hierarchical Self-Assembling Processes of Designed α-Helical Peptides. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003945. [PMID: 33015967 DOI: 10.1002/smll.202003945] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/15/2020] [Indexed: 06/11/2023]
Abstract
Peptide self-assembly is fast evolving into a powerful method for the development of bio-inspired nanomaterials with great potential for many applications, but it remains challenging to control the self-assembling processes and nanostrucutres because of the intricate interplay of various non-covalent interactions. A group of 28-residue α-helical peptides is designed including NN, NK, and HH that display distinct hierarchical events. The key of the design lies in the incorporation of two asparagine (Asn) or histidine (His) residues at the a positions of the second and fourth heptads, which allow one sequence to pack into homodimers with sticky ends through specific interhelical Asn-Asn or metal complexation interactions, followed by their longitudinal association into ordered nanofibers. This is in contrast to classical self-assembling helical peptide systems consisting of two complementary peptides. The collaborative roles played by the four main non-covalent interactions, including hydrogen-bonding, hydrophobic interactions, electrostatic interactions, and metal ion coordination, are well demonstrated during the hierarchical self-assembling processes of these peptides. Different nanostructures, for example, long and short nanofibers, thin and thick fibers, uniform metal ion-entrapped nanofibers, and polydisperse globular stacks, can be prepared by harnessing these interactions at different levels of hierarchy.
Collapse
Affiliation(s)
- Jie Li
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Yurong Zhao
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Peng Zhou
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Xuzhi Hu
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, M13 9PL, UK
| | - Dong Wang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Stephen M King
- ISIS Pulsed Neutron & Muon Source, STFC Rutherford Appleton Laboratory, Didcot, Oxon, OX11 0QX, UK
| | - Sarah E Rogers
- ISIS Pulsed Neutron & Muon Source, STFC Rutherford Appleton Laboratory, Didcot, Oxon, OX11 0QX, UK
| | - Jiqian Wang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao, 266580, China
| | - Jian R Lu
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, M13 9PL, UK
| | - Hai Xu
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao, 266580, China
| |
Collapse
|
22
|
Nagarkar RP, Fichman G, Schneider JP. Engineering and characterization of apH‐sensitive homodimeric antiparallel coiled coil. Pept Sci (Hoboken) 2020. [DOI: 10.1002/pep2.24180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Radhika P. Nagarkar
- Department of Chemistry and Biochemistry University of Delaware Newark Delaware USA
| | - Galit Fichman
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health Frederick Maryland USA
| | - Joel P. Schneider
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health Frederick Maryland USA
| |
Collapse
|
23
|
Kucera J, Terentjev EM. FliI 6-FliJ molecular motor assists with unfolding in the type III secretion export apparatus. Sci Rep 2020; 10:7127. [PMID: 32346005 PMCID: PMC7189227 DOI: 10.1038/s41598-020-63330-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 03/27/2020] [Indexed: 01/05/2023] Open
Abstract
The role of rotational molecular motors of the ATP synthase class is integral to the metabolism of cells. Yet the function of FliI6-FliJ complex, a homolog of the F1 ATPase motor, within the flagellar export apparatus remains unclear. We use a simple two-state model adapted from studies of linear molecular motors to identify key features of this motor. The two states are the 'locked' ground state where the FliJ coiled coil filament experiences angular fluctuations in an asymmetric torsional potential, and a 'free' excited state in which FliJ undergoes rotational diffusion. Michaelis-Menten kinetics was used to treat transitions between these two states, and obtain the average angular velocity of the unloaded FliJ filament within the FliI6 stator: ωmax ≈ 9.0 rps. The motor was then studied under external counter torque conditions in order to ascertain its maximal power output: Pmax ≈ 42 kBT/s (or 102 kW/mol), and the stall torque: Gstall ≈ 3 kBT/rad (or 0.01 nN·nm/rad). Two modes of action within the flagellar export apparatus are proposed, in which the motor performs useful work either by continuously 'grinding' through the resistive environment of the export gate, or by exerting equal and opposite stall force on it. In both cases, the resistance is provided by flagellin subunits entering the flagellar export channel prior to their unfolding. We therefore propose that the function of the FliI6-FliJ complex is to lower the energy barrier, and therefore assist in unfolding of the flagellar proteins before feeding them into the transport channel.
Collapse
Affiliation(s)
- Jiri Kucera
- Cavendish Laboratory, University of Cambridge, Cambridge, CB3 0HE, UK
| | | |
Collapse
|
24
|
Beaulieu ME, Castillo F, Soucek L. Structural and Biophysical Insights into the Function of the Intrinsically Disordered Myc Oncoprotein. Cells 2020; 9:E1038. [PMID: 32331235 PMCID: PMC7226237 DOI: 10.3390/cells9041038] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/13/2022] Open
Abstract
Myc is a transcription factor driving growth and proliferation of cells and involved in the majority of human tumors. Despite a huge body of literature on this critical oncogene, our understanding of the exact molecular determinants and mechanisms that underlie its function is still surprisingly limited. Indubitably though, its crucial and non-redundant role in cancer biology makes it an attractive target. However, achieving successful clinical Myc inhibition has proven challenging so far, as this nuclear protein is an intrinsically disordered polypeptide devoid of any classical ligand binding pockets. Indeed, Myc only adopts a (partially) folded structure in some contexts and upon interacting with some protein partners, for instance when dimerizing with MAX to bind DNA. Here, we review the cumulative knowledge on Myc structure and biophysics and discuss the implications for its biological function and the development of improved Myc inhibitors. We focus this biophysical walkthrough mainly on the basic region helix-loop-helix leucine zipper motif (bHLHLZ), as it has been the principal target for inhibitory approaches so far.
Collapse
Affiliation(s)
| | | | - Laura Soucek
- Peptomyc S.L., Edifici Cellex, 08035 Barcelona, Spain; (F.C.); (L.S.)
- Vall d’Hebron Institute of Oncology (VHIO), Edifici Cellex, 08035 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08035 Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08035 Bellaterra, Spain
| |
Collapse
|
25
|
Sinha NJ, Wu D, Kloxin CJ, Saven JG, Jensen GV, Pochan DJ. Polyelectrolyte character of rigid rod peptide bundlemer chains constructed via hierarchical self-assembly. SOFT MATTER 2019; 15:9858-9870. [PMID: 31738361 DOI: 10.1039/c9sm01894h] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Short α-helical peptides were computationally designed to self-assemble into robust coiled coils that are antiparallel, homotetrameric bundles. These peptide bundle units, or 'bundlemers', have been utilized as anisotropic building blocks to construct bundlemer-based polymers via a hierarchical, hybrid physical-covalent assembly pathway. The bundlemer chains were constructed using short linker connections via 'click' chemistry reactions between the N-termini of bundlemer constituent peptides. The resulting bundlemer chains appear as extremely rigid, cylindrical rods in transmission electron microscopy (TEM) images. Small angle neutron scattering (SANS) shows that these bundlemer chains exist as individual rods in solution with a cross-section that is equal to that of a single coiled coil bundlemer building block of ≈20 Å. SANS further confirms that the interparticle solution structure of the rigid rod bundlemer chains is heterogeneous and responsive to solution conditions, such as ionic-strength and pH. Due to their peptidic constitution, the bundlemer assemblies behave like polyelectrolytes that carry an average charge density of approximately 3 charges per bundlemer as determined from SANS structure factor data fitting, which describes the repulsion between charged rods in solution. This repulsion manifests as a correlation hole in the scattering profile that is suppressed by dilution or addition of salt. Presence of rod cluster aggregates with a mass fractal dimension of ≈2.5 is also confirmed across all samples. The formation of such dense, fractal-like cluster aggregates in a solution of net repulsive rods is a unique example of the subtle balance between short-range attraction and long-rage repulsion interactions in proteins and other biomaterials. With computational control of constituent peptide sequences, it is further possible to deconvolute the underlying sequence driven structure-property relationships in the modular bundlemer chains.
Collapse
Affiliation(s)
- Nairiti J Sinha
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, USA.
| | | | | | | | | | | |
Collapse
|
26
|
Endoplasmic reticulum retention signaling and transmembrane channel proteins predicted for oilseed ω3 fatty acid desaturase 3 (FAD3) genes. Funct Integr Genomics 2019; 20:433-458. [PMID: 31781992 DOI: 10.1007/s10142-019-00718-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 09/19/2019] [Accepted: 09/23/2019] [Indexed: 10/25/2022]
Abstract
Oilseed crop oils contain a variety of unsaturated fatty acids that are synthesized and regulated by fatty acid desaturases (FADs). In this study, 14 FAD3 (ω3 desaturase) protein sequences from oilseeds are analyzed and presented through the application of several computational tools. The results indicated a close relationship between Brassica napus and Camelina sativa, as well as between Salvia hispanica and Perilla frutescens FAD3s, due to a high similarity in codon preferences in codon usage clusters and the phylogenetic tree. The cis-acting element results reveal that the seed-specific promoter region of BnFAD3 contains the critical conserved boxes such as HSE and ABRE, which are involved in responsiveness to heat stress and abscisic acid. The presence of the aforementioned conserved boxes may increase cold acclimation as well as tolerance to drought and high salinity. Omega(ω)3 desaturases contain a Skn-1 motif which is a cis-acting regulatory element required involved in endosperm development. In oilseed FAD3s, leucine is the most repeated amino acid in FAD3 proteins. The study conveyed that B. napus, Camelina sativa, Linum usitatissimum, Vernicia fordii, Gossypium hirsutum, S. hispanica, Cannabis sativa, and P. frutescens have retention signal KXKXX/XKXX at their c-terminus sites, which is one of the most important characteristics of FADs. Additionally, it was found that BnFAD3 is a transmembrane protein that can convert ω6 to ω3 fatty acids and may simultaneously act as a potassium ion channel in the ER.
Collapse
|
27
|
Grayson KJ, Anderson JLR. Designed for life: biocompatible de novo designed proteins and components. J R Soc Interface 2019; 15:rsif.2018.0472. [PMID: 30158186 PMCID: PMC6127164 DOI: 10.1098/rsif.2018.0472] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/01/2018] [Indexed: 12/30/2022] Open
Abstract
A principal goal of synthetic biology is the de novo design or redesign of biomolecular components. In addition to revealing fundamentally important information regarding natural biomolecular engineering and biochemistry, functional building blocks will ultimately be provided for applications including the manufacture of valuable products and therapeutics. To fully realize this ambitious goal, the designed components must be biocompatible, working in concert with natural biochemical processes and pathways, while not adversely affecting cellular function. For example, de novo protein design has provided us with a wide repertoire of structures and functions, including those that can be assembled and function in vivo. Here we discuss such biocompatible designs, as well as others that have the potential to become biocompatible, including non-protein molecules, and routes to achieving full biological integration.
Collapse
Affiliation(s)
- Katie J Grayson
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, Bristol BS8 1TD, UK
| | - J L Ross Anderson
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, Bristol BS8 1TD, UK .,BrisSynBio Synthetic Biology Research Centre, University of Bristol, Life Sciences Building, Tyndall Avenue, Bristol BS8 1TQ, UK
| |
Collapse
|
28
|
Rhys GG, Wood CW, Beesley JL, Zaccai NR, Burton AJ, Brady RL, Thomson AR, Woolfson DN. Navigating the Structural Landscape of De Novo α-Helical Bundles. J Am Chem Soc 2019; 141:8787-8797. [PMID: 31066556 DOI: 10.1021/jacs.8b13354] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The association of amphipathic α helices in water leads to α-helical-bundle protein structures. However, the driving force for this-the hydrophobic effect-is not specific and does not define the number or the orientation of helices in the associated state. Rather, this is achieved through deeper sequence-to-structure relationships, which are increasingly being discerned. For example, for one structurally extreme but nevertheless ubiquitous class of bundle-the α-helical coiled coils-relationships have been established that discriminate between all-parallel dimers, trimers, and tetramers. Association states above this are known, as are antiparallel and mixed arrangements of the helices. However, these alternative states are less well understood. Here, we describe a synthetic-peptide system that switches between parallel hexamers and various up-down-up-down tetramers in response to single-amino-acid changes and solution conditions. The main accessible states of each peptide variant are characterized fully in solution and, in most cases, to high resolution with X-ray crystal structures. Analysis and inspection of these structures helps rationalize the different states formed. This navigation of the structural landscape of α-helical coiled coils above the dimers and trimers that dominate in nature has allowed us to design rationally a well-defined and hyperstable antiparallel coiled-coil tetramer (apCC-Tet). This robust de novo protein provides another scaffold for further structural and functional designs in protein engineering and synthetic biology.
Collapse
Affiliation(s)
- Guto G Rhys
- School of Chemistry , University of Bristol , Cantock's Close , Bristol BS8 1TS , United Kingdom
| | - Christopher W Wood
- School of Chemistry , University of Bristol , Cantock's Close , Bristol BS8 1TS , United Kingdom
| | - Joseph L Beesley
- School of Chemistry , University of Bristol , Cantock's Close , Bristol BS8 1TS , United Kingdom
| | - Nathan R Zaccai
- School of Biochemistry , University of Bristol , Medical Sciences Building, University Walk , Bristol BS8 1TD , United Kingdom
| | - Antony J Burton
- School of Chemistry , University of Bristol , Cantock's Close , Bristol BS8 1TS , United Kingdom
- Frick Chemistry Laboratory , Princeton University , Princeton , New Jersey 08544 , United States
| | - R Leo Brady
- School of Biochemistry , University of Bristol , Medical Sciences Building, University Walk , Bristol BS8 1TD , United Kingdom
| | - Andrew R Thomson
- School of Chemistry , University of Bristol , Cantock's Close , Bristol BS8 1TS , United Kingdom
- School of Chemistry , University of Glasgow , Glasgow G12 8QQ , United Kingdom
| | - Derek N Woolfson
- School of Chemistry , University of Bristol , Cantock's Close , Bristol BS8 1TS , United Kingdom
- School of Biochemistry , University of Bristol , Medical Sciences Building, University Walk , Bristol BS8 1TD , United Kingdom
- BrisSynBio , University of Bristol , Life Sciences Building, Tyndall Avenue , Bristol BS8 1TQ , United Kingdom
| |
Collapse
|
29
|
Rahman MM, Ueda M, Hirose T, Ito Y. Spontaneous Formation of Gating Lipid Domain in Uniform-Size Peptide Vesicles for Controlled Release. J Am Chem Soc 2018; 140:17956-17961. [PMID: 30525544 DOI: 10.1021/jacs.8b09362] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Hybrid assemblies composed of phospholipids and amphiphilic polymers have been investigated previously as a biomimetic model of biological cells. However, these studies focused on the functions of polymers in a sea of membrane lipids. Here, we prepared a highly stable peptide-lipid hybrid vesicle from a combination of an amphiphilic polypeptide and the phospholipid, 1,2-dimyristoyl- sn-glycero-3-phosphocholine, with a mixing molar ratio of 1:1. The phase-separated structure of the hybrid vesicle was demonstrated by fluorescence resonance energy transfer analysis. The lipid domain of the hybrid vesicle had a phase-transition temperature of 38 °C and allowed the permeation of a hydrophilic molecule, fluorescein isothiocyanate-labeled polyethylene glycol ( Mw: 2000), above 38 °C. The designed peptide-lipid hybrid vesicle and a "lipidic gate" are a promising tool for smart drug delivery.
Collapse
Affiliation(s)
- Md Mofizur Rahman
- Emergent Bioengineering Materials Research Team , RIKEN Center for Emergent Matter Science (CEMS) , 2-1 Hirosawa, Wako , Saitama 351-0198 , Japan.,Graduate School of Science and Engineering , Saitama University , 255 Shimo-Okubo, Sakura-ku, Saitama City , Saitama 338-8570 , Japan
| | - Motoki Ueda
- Emergent Bioengineering Materials Research Team , RIKEN Center for Emergent Matter Science (CEMS) , 2-1 Hirosawa, Wako , Saitama 351-0198 , Japan.,Nano Medical Engineering Laboratory , RIKEN Cluster for Pioneering Research (CPR) , 2-1 Hirosawa, Wako , Saitama 351-0198 , Japan
| | - Takuji Hirose
- Graduate School of Science and Engineering , Saitama University , 255 Shimo-Okubo, Sakura-ku, Saitama City , Saitama 338-8570 , Japan
| | - Yoshihiro Ito
- Emergent Bioengineering Materials Research Team , RIKEN Center for Emergent Matter Science (CEMS) , 2-1 Hirosawa, Wako , Saitama 351-0198 , Japan.,Nano Medical Engineering Laboratory , RIKEN Cluster for Pioneering Research (CPR) , 2-1 Hirosawa, Wako , Saitama 351-0198 , Japan
| |
Collapse
|
30
|
Effect of l -histidine on the heat-induced aggregation of bighead carp ( Aristichthys nobilis ) myosin in low/high ionic strength solution. Food Hydrocoll 2018. [DOI: 10.1016/j.foodhyd.2017.08.029] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
31
|
Xu X, Xiao X, Xu S, Liu H. Computational insights into the destabilization of α-helical conformations formed by leucine zipper peptides in response to temperature. Phys Chem Chem Phys 2018; 18:25465-25473. [PMID: 27722604 DOI: 10.1039/c6cp05145f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Recent experiments in our lab (Phys. Chem. Chem. Phys., 2016, 18, 10129-10137) suggested using leucine zipper peptides to enhance the thermosensitivity of liposomes. To understand the mechanisms of temperature-responsive control by the leucine zipper peptide in liposomes, we firstly performed quantum mechanics calculations and implicit-solvent replica exchange molecular dynamics simulations to study the thermo-stability of two leucine zipper peptides, CH3(CH2)4-CO-[VAQLEVK-VAQLESK-VSKLESK-VSSLESK] (termed the capped peptide) and A-[VAQLEVK-VAQLESK-VSKLESK-VSSLESK] (termed the ALA peptide). The analysis of dihedral angle principal components and protein secondary structures was conducted to determine the temperature-dependence conformation transition of the two peptides. Simulation results revealed that our computed transition temperature of the capped peptide is 319.1 K that accords with experimental measurement, 321.1 K. Later, explicit-solvent conventional molecular dynamics simulations were carried out to examine the process of folding and unfolding of the ALA and capped peptides complexed with a lipid bilayer and water in the vicinity of their transition temperatures. A further analysis of conformation and energy of the folded peptides showed that the increase of temperature gives rise to a notable decrease in the number of intra-chain hydrogen bonds and a significant increase in the potential energy of the peptides, thereby reducing the folding stability of the two peptides. As compared to the ALA peptide, a lower transition temperature caused by less intra-chain hydrogen bonds was observed in the capped peptide, which is closer to the temperature of tumor cells. This fact suggests that the capped peptide is more suitable to produce highly sensitive liposomes for the delivery of cancer drugs.
Collapse
Affiliation(s)
- Xiejun Xu
- State Key Laboratory of Chemical Engineering and School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| | - Xingqing Xiao
- Chemical and Biomolecular Engineering Department, North Carolina State University, Raleigh, North Carolina 27695-7905, USA
| | - Shouhong Xu
- State Key Laboratory of Chemical Engineering and School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| | - Honglai Liu
- State Key Laboratory of Chemical Engineering and School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
32
|
Bhattacharjee P, Dasgupta D, Sengupta K. DCM associated LMNA mutations cause distortions in lamina structure and assembly. Biochim Biophys Acta Gen Subj 2017; 1861:2598-2608. [PMID: 28844980 DOI: 10.1016/j.bbagen.2017.08.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/01/2017] [Accepted: 08/11/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND A and B-type lamins are integral scaffolding components of the nuclear lamina which impart rigidity and shape to all metazoan nuclei. Over 450 mutations in A-type lamins are associated with 16 human diseases including dilated cardiomyopathy (DCM). Here, we show that DCM mutants perturb the self-association of lamin A (LA) and it's binding with lamin B1 (LB1). METHODS We used confocal and superresolution microscopy (NSIM) to study the effect of LA mutants on the nuclear lamina. We further used circular dichroism, fluorescence spectroscopy and isothermal titration calorimetry (ITC) to probe the structural modulations, self-association and heteropolymeric association of mutant LA. RESULTS Transfection of mutants in cultured cell lines result in the formation of nuclear aggregates of varied size and distribution. Endogenous LB1 is sequestered into these aggregates. This is consistent with the ten-fold increase in association constant of the mutant proteins compared to the wild type. These mutants exhibit differential heterotypic interaction with LB1, along with significant secondary and tertiary structural alterations of the interacting proteins. Thermodynamic studies demonstrate that the mutants bind to LB1 with different stoichiometry, affinity and energetics. CONCLUSIONS In this report we show that increased self-association propensity of mutant LA modulates the LA-LB1 interaction and precludes the formation of an otherwise uniform laminar network. GENERAL SIGNIFICANCE Our results might highlight the role of homotypic and heterotypic interactions of LA in the pathogenesis of DCM and hence laminopathies in the broader sense.
Collapse
Affiliation(s)
- Pritha Bhattacharjee
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Dipak Dasgupta
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India.
| | - Kaushik Sengupta
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India.
| |
Collapse
|
33
|
Wang HF, Wibowo D, Shao Z, Middelberg APJ, Zhao CX. Design of Modular Peptide Surfactants and Their Surface Activity. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:7957-7967. [PMID: 28732169 DOI: 10.1021/acs.langmuir.7b01382] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Designed peptide surfactants offer a number of advanced properties over conventional petrochemical surfactants, including biocompatibility, sustainability, and tailorability of the chemical and physical properties through peptide design. Their biocompatibility and degradability make them attractive for various applications, particularly for food and pharmaceutical applications. In this work, two new peptide surfactants derived from an amphiphilic peptide surfactant (AM1) were designed (AM-S and C8-AM) to better understand links between structure, interfacial activity, and emulsification. Based on AM1, which has an interfacial α-helical structure, AM-S and C8-AM were designed to have two modules, that is, the α-helical AM1 module and an additional hydrophobic moiety to provide for better anchoring at the oil-water interface. Both AM-S and C8-AM at low bulk concentration of 20 μM were able to adsorb rapidly at the oil-water interface and reduced interfacial tension to equilibrium values of 17.0 and 8.4 mN/m within 400 s, respectively. Their relatively quick adsorption kinetics allowed the formation of nanoemulsions with smaller droplet sizes and narrower size distribution. AM-S and C8-AM at 800 μM bulk concentration could make nanoemulsions of average diameters 180 and 147 nm, respectively, by simple sonication. With respect to the long-term stability, a minimum peptide concentration of 400 μM for AM-S and a lower concentration of 100 μM for C8-AM were demonstrated to effectively stabilize nanoemulsions over 3 weeks. Compared to AM1, the AM-S nanoemulsion retained its stimuli-responsive function triggered by metal ions, whereas the C8-AM nanoemulsions did not respond to the stimuli as efficiently as AM-S because of the strong anchoring ability of the hydrophobic C8 module. The two-module design of AM-S and C8-AM represents a new strategy in tuning the surface activity of peptide surfactants, offering useful information and guidance of future designs.
Collapse
Affiliation(s)
- Hao-Fei Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , St Lucia QLD 4072, Australia
| | - David Wibowo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , St Lucia QLD 4072, Australia
| | - Zhengzhong Shao
- State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials and Department of Macromolecular Science, Fudan University , Shanghai 200433, China
| | - Anton P J Middelberg
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , St Lucia QLD 4072, Australia
| | - Chun-Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , St Lucia QLD 4072, Australia
| |
Collapse
|
34
|
Fibre diffraction studies of biological macromolecules. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 127:43-87. [DOI: 10.1016/j.pbiomolbio.2017.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/21/2017] [Accepted: 04/05/2017] [Indexed: 12/27/2022]
|
35
|
Pulido D, Hussain SA, Hohenester E. Crystal Structure of the Heterotrimeric Integrin-Binding Region of Laminin-111. Structure 2017; 25:530-535. [PMID: 28132784 PMCID: PMC5343747 DOI: 10.1016/j.str.2017.01.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 12/16/2016] [Accepted: 01/05/2017] [Indexed: 01/05/2023]
Abstract
Laminins are cell-adhesive glycoproteins that are essential for basement membrane assembly and function. Integrins are important laminin receptors, but their binding site on the heterotrimeric laminins is poorly defined structurally. We report the crystal structure at 2.13 Å resolution of a minimal integrin-binding fragment of mouse laminin-111, consisting of ∼50 residues of α1β1γ1 coiled coil and the first three laminin G-like (LG) domains of the α1 chain. The LG domains adopt a triangular arrangement, with the C terminus of the coiled coil situated between LG1 and LG2. The critical integrin-binding glutamic acid residue in the γ1 chain tail is surface exposed and predicted to bind to the metal ion-dependent adhesion site in the integrin β1 subunit. Additional contacts to the integrin are likely to be made by the LG1 and LG2 surfaces adjacent to the γ1 chain tail, which are notably conserved and free of obstructing glycans.
Collapse
Affiliation(s)
- David Pulido
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, London SW7 2AZ, UK
| | - Sadaf-Ahmahni Hussain
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, London SW7 2AZ, UK
| | - Erhard Hohenester
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, London SW7 2AZ, UK.
| |
Collapse
|
36
|
Schmidt NW, Grigoryan G, DeGrado WF. The accommodation index measures the perturbation associated with insertions and deletions in coiled-coils: Application to understand signaling in histidine kinases. Protein Sci 2017; 26:414-435. [PMID: 27977891 PMCID: PMC5326573 DOI: 10.1002/pro.3095] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 11/30/2016] [Accepted: 12/01/2016] [Indexed: 01/08/2023]
Abstract
Coiled-coils are essential components of many protein complexes. First discovered in structural proteins such as keratins, they have since been found to figure largely in the assembly and dynamics required for diverse functions, including membrane fusion, signal transduction and motors. Coiled-coils have a characteristic repeating seven-residue geometric and sequence motif, which is sometimes interrupted by the insertion of one or more residues. Such insertions are often highly conserved and critical to interdomain communication in signaling proteins such as bacterial histidine kinases. Here we develop the "accommodation index" as a parameter that allows automatic detection and classification of insertions based on the three dimensional structure of a protein. This method allows precise identification of the type of insertion and the "accommodation length" over which the insertion is structurally accommodated. A simple theory is presented that predicts the structural perturbations of 1, 3, 4 residue insertions as a function of the length over which the insertion is accommodated. Analysis of experimental structures is in good agreement with theory, and shows that short accommodation lengths give rise to greater perturbation of helix packing angles, changes in local helical phase, and increased structural asymmetry relative to long accommodation lengths. Cytoplasmic domains of histidine kinases in different signaling states display large changes in their accommodation lengths, which can now be seen to underlie diverse structural transitions including symmetry/asymmetry and local variations in helical phase that accompany signal transduction.
Collapse
Affiliation(s)
- Nathan W. Schmidt
- Department of Pharmaceutical ChemistryCardiovascular Research Institute, University of CaliforniaSan FranciscoCalifornia94158
| | - Gevorg Grigoryan
- Department of Computer ScienceDartmouth CollegeHanoverNew Hampshire03755
- Department of Biological SciencesDartmouth CollegeHanoverNew Hampshire03755
| | - William F. DeGrado
- Department of Pharmaceutical ChemistryCardiovascular Research Institute, University of CaliforniaSan FranciscoCalifornia94158
| |
Collapse
|
37
|
Klement M, Zheng J, Liu C, Tan HL, Wong VVT, Choo ABH, Lee DY, Ow DSW. Antibody engineering of a cytotoxic monoclonal antibody 84 against human embryonic stem cells: Investigating the effects of multivalency on cytotoxicity. J Biotechnol 2017; 243:29-37. [PMID: 28042013 DOI: 10.1016/j.jbiotec.2016.12.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 11/22/2016] [Accepted: 12/27/2016] [Indexed: 02/06/2023]
Abstract
Antibody fragments have shown targeted specificity to their antigens, but only modest tissue retention times in vivo and in vitro. Multimerization has been used as a protein engineering tool to increase the number of binding units and thereby enhance the efficacy and retention time of antibody fragments. In this work, we explored the effects of valency using a series of self-assembling polypeptides based on the GCN4 leucine zipper multimerization domain fused to a single-chain variable fragment via an antibody upper hinge sequence. Four engineered antibody fragments with a valency from one to four antigen-binding units of a cytotoxic monoclonal antibody 84 against human embryonic stem cells (hESC) were constructed. We hypothesized that higher cytotoxicity would be observed for fragments with increased valency. Flow cytometry analysis revealed that the trimeric and tetrameric engineered antibody fragments resulted in the highest degree of cytotoxicity to the undifferentiated hESC, while the engineered antibody fragments were observed to have improved tissue penetration into cell clusters. Thus, a trade off was made for the trimeric versus tetrameric fragment due to improved tissue penetration. These results have direct implications for antibody-mediated removal of undifferentiated hESC during regenerative medicine and cell therapy.
Collapse
Affiliation(s)
- Maximilian Klement
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore; Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117576, Singapore
| | - Jiyun Zheng
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, #05-01, 117456, Singapore
| | - Chengcheng Liu
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore
| | - Heng-Liang Tan
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore
| | - Victor Vai Tak Wong
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore
| | - Andre Boon-Hwa Choo
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore; Department of Biomedical Engineering, National University of Singapore, 9 Engineering Drive 1, 117575, Singapore
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore; Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117576, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, 117456, Singapore.
| | - Dave Siak-Wei Ow
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01 Centros, 138668, Singapore.
| |
Collapse
|
38
|
Abstract
α-Helical coiled coils constitute one of the most diverse folds yet described. They range in length over two orders of magnitude; they form rods, segmented ropes, barrels, funnels, sheets, spirals, and rings, which encompass anywhere from two to more than 20 helices in parallel or antiparallel orientation; they assume different helix crossing angles, degrees of supercoiling, and packing geometries. This structural diversity supports a wide range of biological functions, allowing them to form mechanically rigid structures, provide levers for molecular motors, project domains across large distances, mediate oligomerization, transduce conformational changes and facilitate the transport of other molecules. Unlike almost any other protein fold known to us, their structure can be computed from parametric equations, making them an ideal model system for rational protein design. Here we outline the principles by which coiled coils are structured, review the determinants of their folding and stability, and present an overview of their diverse architectures.
Collapse
|
39
|
Abstract
Fibrinogen and fibrin are essential for hemostasis and are major factors in thrombosis, wound healing, and several other biological functions and pathological conditions. The X-ray crystallographic structure of major parts of fibrin(ogen), together with computational reconstructions of missing portions and numerous biochemical and biophysical studies, have provided a wealth of data to interpret molecular mechanisms of fibrin formation, its organization, and properties. On cleavage of fibrinopeptides by thrombin, fibrinogen is converted to fibrin monomers, which interact via knobs exposed by fibrinopeptide removal in the central region, with holes always exposed at the ends of the molecules. The resulting half-staggered, double-stranded oligomers lengthen into protofibrils, which aggregate laterally to make fibers, which then branch to yield a three-dimensional network. Much is now known about the structural origins of clot mechanical properties, including changes in fiber orientation, stretching and buckling, and forced unfolding of molecular domains. Studies of congenital fibrinogen variants and post-translational modifications have increased our understanding of the structure and functions of fibrin(ogen). The fibrinolytic system, with the zymogen plasminogen binding to fibrin together with tissue-type plasminogen activator to promote activation to the active proteolytic enzyme, plasmin, results in digestion of fibrin at specific lysine residues. In spite of a great increase in our knowledge of all these interconnected processes, much about the molecular mechanisms of the biological functions of fibrin(ogen) remains unknown, including some basic aspects of clotting, fibrinolysis, and molecular origins of fibrin mechanical properties. Even less is known concerning more complex (patho)physiological implications of fibrinogen and fibrin.
Collapse
Affiliation(s)
- John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
40
|
Abstract
During the 1930s and 1940s the technique of X-ray diffraction was applied widely by William Astbury and his colleagues to a number of naturally-occurring fibrous materials. On the basis of the diffraction patterns obtained, he observed that the structure of each of the fibres was dominated by one of a small number of different types of molecular conformation. One group of fibres, known as the k-m-e-f group of proteins (keratin - myosin - epidermin - fibrinogen), gave rise to diffraction characteristics that became known as the α-pattern. Others, such as those from a number of silks, gave rise to a different pattern - the β-pattern, while connective tissues yielded a third unique set of diffraction characteristics. At the time of Astbury's work, the structures of these materials were unknown, though the spacings of the main X-ray reflections gave an idea of the axial repeats and the lateral packing distances. In a breakthrough in the early 1950s, the basic structures of all of these fibrous proteins were determined. It was found that the long protein chains, composed of strings of amino acids, could be folded up in a systematic manner to generate a limited number of structures that were consistent with the X-ray data. The most important of these were known as the α-helix, the β-sheet, and the collagen triple helix. These studies provided information about the basic building blocks of all proteins, both fibrous and globular. They did not, however, provide detailed information about how these molecules packed together in three-dimensions to generate the fibres found in vivo. A number of possible packing arrangements were subsequently deduced from the X-ray diffraction and other data, but it is only in the last few years, through the continued improvements of electron microscopy, that the packing details within some fibrous proteins can now be seen directly. Here we outline briefly some of the milestones in fibrous protein structure determination, the role of the amino acid sequences and how new techniques, including electron microscopy, are helping to define fibrous protein structures in three-dimensions. We also introduce the idea that, from the known sequence characteristics of different fibrous proteins, new molecules can be designed and synthesized, thereby generating new biological materials with specific structural properties. Some of these, for example, are planned for use in drug delivery systems. Along the way we also introduce the various Chapters of the book, where individual fibrous proteins are discussed in detail.
Collapse
|
41
|
Kefala A, Kotsifaki D, Providaki M, Amprazi M, Kokkinidis M. Expression, purification and crystallization of a protein resulting from the inversion of the amino-acid sequence of a helical bundle. Acta Crystallogr F Struct Biol Commun 2017; 73:51-53. [PMID: 28045394 PMCID: PMC5287371 DOI: 10.1107/s2053230x16020173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 12/19/2016] [Indexed: 11/10/2022] Open
Abstract
Earlier studies have found that the occurrence of inverse sequence identity in proteins is not indicative of three-dimensional similarity, but rather leads to different folds or unfolded proteins. Short helices, however, frequently keep their conformations when their sequences are inverted. To explore the impact of sequence inversion on long helices, revRM6, with the inverse amino-acid sequence relative to RM6, a highly stable variant of the ColE1 Rop protein, was engineered. RM6 is a highly regular four-α-helical bundle that serves as a model system for protein-folding studies. Here, the crystallization and preliminary crystallographic characterization of revRM6 are reported. The protein was overexpressed in Escherichia coli, purified to homogeneity and crystallized. The crystals belonged to space group P41212, with unit-cell parameters a = b = 44.98, c = 159.74 Å, and diffracted to a resolution of 3.45 Å.
Collapse
Affiliation(s)
- Aikaterini Kefala
- Department of Biology, University of Crete, Voutes University Campus, PO Box 2208, 70013 Heraklion, Crete, Greece
| | - Dina Kotsifaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology – Hellas, Nikolaou Plastira Street 100, 70013 Heraklion, Crete, Greece
| | - Mary Providaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology – Hellas, Nikolaou Plastira Street 100, 70013 Heraklion, Crete, Greece
| | - Maria Amprazi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology – Hellas, Nikolaou Plastira Street 100, 70013 Heraklion, Crete, Greece
| | - Michael Kokkinidis
- Department of Biology, University of Crete, Voutes University Campus, PO Box 2208, 70013 Heraklion, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology – Hellas, Nikolaou Plastira Street 100, 70013 Heraklion, Crete, Greece
| |
Collapse
|
42
|
Wu Y, Collier JH. α-Helical coiled-coil peptide materials for biomedical applications. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [PMID: 27597649 DOI: 10.1002/wnan.1424] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/07/2016] [Accepted: 07/17/2016] [Indexed: 12/31/2022]
Abstract
Self-assembling coiled coils, which occur commonly in native proteins, have received significant interest for the design of new biomaterials-based medical therapies. Considerable effort over recent years has led to a detailed understanding of the self-assembly process of coiled coils, and a diverse collection of strategies have been developed for designing functional materials using this motif. The ability to engineer the interface between coiled coils allows one to achieve variously connected components, leading to precisely defined structures such as nanofibers, nanotubes, nanoparticles, networks, gels, and combinations of these. Currently these materials are being developed for a range of biotechnological and medical applications, including drug delivery systems for controlled release, targeted nanomaterials, 'drug-free' therapeutics, vaccine delivery systems, and others. WIREs Nanomed Nanobiotechnol 2017, 9:e1424. doi: 10.1002/wnan.1424 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Yaoying Wu
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
43
|
McGrath KP, Butler MM, DiGirolamo CM, Kaplan DL, Petka WA, Laue TM. Electrostatic Interactions in Leucine Zippers: Effects on Stability and Specificity of Interaction. J BIOACT COMPAT POL 2016. [DOI: 10.1177/088391150001500405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Using a set of natural coiled-coil proteins as models, a series of recombinant proteins were designed and expressed in E. coli. These proteins contain a consensus coiled-coil sequence as a framework into which were incorporated positively or negatively charged residues at selected positions. A mixed-site genetic strategy was used to generate DNA fragments encoding over 4,000 different combinations of charged residues within the coiled-coil motif. A subset of these genes was used to produce recombinant coiled-coil proteins with well-defined variations in charge pattern and composition. Variants of each sequence containing a unique cysteine at the C-terminus were oxidized to the disulfide-linked dimer, and characterization of their physical properties support the proposed parallel orientation of protein chains. In all cases, equilibrium populations of dimeric and tetrameric structures were observed under physiological conditions, with dimer-to-tetramer dissociation constants in the range of 50-190 riM. Significant differences in complex stability were seen with different charge patterns. Contrary to expectations, no linear relationship was observed between net ionic interaction and any measure of complex stability, arguing that a more subtle set of rules governs these interactions. This work has revealed two important aspects of coiled-coil interactions: the observed relationship between charge interactions and complex stability shows considerable nonlinearity, and the presence of higher order interactions in coiled-coil motifs may be more widespread than is currently suspected. The relationships described here have broad relevance, especially in the areas of protein folding, protein-based materials design, antibody-antigen and receptor-ligand interactions, and rational drug design.
Collapse
Affiliation(s)
- Kevin P. McGrath
- Science and Technology Directorate, U.S. Army Natick RD&E Center, Kansas St., Natick, MA 01760-5020
| | - Michelle M. Butler
- Science and Technology Directorate, U.S. Army Natick RD&E Center, Kansas St., Natick, MA 01760-5020
| | - Carla M. DiGirolamo
- Science and Technology Directorate, U.S. Army Natick RD&E Center, Kansas St., Natick, MA 01760-5020
| | - David L. Kaplan
- Science and Technology Directorate, U.S. Army Natick RD&E Center, Kansas St., Natick, MA 01760-5020
| | - Wendy A. Petka
- Polymer Science and Engineering, University of Massachusetts, Amherst, MA 01003
| | - Thomas M. Laue
- Dept. of Biochemistry, University of New Hampshire, Durham, NH 03824
| |
Collapse
|
44
|
Kulish O, Wright AD, Terentjev EM. F1 rotary motor of ATP synthase is driven by the torsionally-asymmetric drive shaft. Sci Rep 2016; 6:28180. [PMID: 27321713 PMCID: PMC4913325 DOI: 10.1038/srep28180] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/31/2016] [Indexed: 11/26/2022] Open
Abstract
F1F0 ATP synthase (ATPase) either facilitates the synthesis of ATP in a process driven by the proton moving force (pmf), or uses the energy from ATP hydrolysis to pump protons against the concentration gradient across the membrane. ATPase is composed of two rotary motors, F0 and F1, which compete for control of their shared γ -shaft. We present a self-consistent physical model of F1 motor as a simplified two-state Brownian ratchet using the asymmetry of torsional elastic energy of the coiled-coil γ -shaft. This stochastic model unifies the physical concepts of linear and rotary motors, and explains the stepped unidirectional rotary motion. Substituting the model parameters, all independently known from recent experiments, our model quantitatively reproduces the ATPase operation, e.g. the ‘no-load’ angular velocity is ca. 400 rad/s anticlockwise at 4 mM ATP. Increasing the pmf torque exerted by F0 can slow, stop and overcome the torque generated by F1, switching from ATP hydrolysis to synthesis at a very low value of ‘stall torque’. We discuss the motor efficiency, which is very low if calculated from the useful mechanical work it produces - but is quite high when the ‘useful outcome’ is measured in the number of H+ pushed against the chemical gradient.
Collapse
Affiliation(s)
- O Kulish
- Cavendish Laboratory, University of Cambridge, Cambridge CB3 0HE, UK
| | - A D Wright
- Cavendish Laboratory, University of Cambridge, Cambridge CB3 0HE, UK
| | - E M Terentjev
- Cavendish Laboratory, University of Cambridge, Cambridge CB3 0HE, UK
| |
Collapse
|
45
|
Krieg E, Bastings MMC, Besenius P, Rybtchinski B. Supramolecular Polymers in Aqueous Media. Chem Rev 2016; 116:2414-77. [DOI: 10.1021/acs.chemrev.5b00369] [Citation(s) in RCA: 570] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
| | | | - Pol Besenius
- Institute
of Organic Chemistry, Johannes Gutenberg-Universität Mainz, Mainz 55128, Germany
| | - Boris Rybtchinski
- Department
of Organic Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
46
|
Belnap DM. Electron Microscopy and Image Processing: Essential Tools for Structural Analysis of Macromolecules. ACTA ACUST UNITED AC 2015; 82:17.2.1-17.2.61. [PMID: 26521712 DOI: 10.1002/0471140864.ps1702s82] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Macromolecular electron microscopy typically depicts the structures of macromolecular complexes ranging from ∼200 kDa to hundreds of MDa. The amount of specimen required, a few micrograms, is typically 100 to 1000 times less than needed for X-ray crystallography or nuclear magnetic resonance spectroscopy. Micrographs of frozen-hydrated (cryogenic) specimens portray native structures, but the original images are noisy. Computational averaging reduces noise, and three-dimensional reconstructions are calculated by combining different views of free-standing particles ("single-particle analysis"). Electron crystallography is used to characterize two-dimensional arrays of membrane proteins and very small three-dimensional crystals. Under favorable circumstances, near-atomic resolutions are achieved. For structures at somewhat lower resolution, pseudo-atomic models are obtained by fitting high-resolution components into the density. Time-resolved experiments describe dynamic processes. Electron tomography allows reconstruction of pleiomorphic complexes and subcellular structures and modeling of macromolecules in their cellular context. Significant information is also obtained from metal-coated and dehydrated specimens.
Collapse
Affiliation(s)
- David M Belnap
- Departments of Biology and Biochemistry, University of Utah, Salt Lake City, Utah
| |
Collapse
|
47
|
Key Residues of Outer Membrane Protein OprI Involved in Hexamer Formation and Bacterial Susceptibility to Cationic Antimicrobial Peptides. Antimicrob Agents Chemother 2015; 59:6210-22. [PMID: 26248382 DOI: 10.1128/aac.01406-15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 07/17/2015] [Indexed: 11/20/2022] Open
Abstract
Antimicrobial peptides (AMPs) are important components of the host innate defense mechanism against invading pathogens. Our previous studies have shown that the outer membrane protein, OprI from Pseudomonas aeruginosa or its homologue, plays a vital role in the susceptibility of Gram-negative bacteria to cationic α-helical AMPs (Y. M. Lin, S. J. Wu, T. W. Chang, C. F. Wang, C. S. Suen, M. J. Hwang, M. D. Chang, Y. T. Chen, Y. D. Liao, J Biol Chem 285:8985-8994, 2010, http://dx.doi.org/10.1074/jbc.M109.078725; T. W. Chang, Y. M. Lin, C. F. Wang, Y. D. Liao, J Biol Chem 287:418-428, 2012, http://dx.doi.org/10.1074/jbc.M111.290361). Here, we obtained two forms of recombinant OprI: rOprI-F, a hexamer composed of three disulfide-bridged dimers, was active in AMP binding, while rOprI-R, a trimer, was not. All the subunits predominantly consisted of α-helices and exhibited rigid structures with a melting point centered around 76°C. Interestingly, OprI tagged with Escherichia coli signal peptide was expressed in a hexamer, which was anchored on the surface of E. coli, possibly through lipid acids added at the N terminus of OprI and involved in the binding and susceptibility to AMP as native P. aeruginosa OprI. Deletion and mutation studies showed that Cys1 and Asp27 played a key role in hexamer formation and AMP binding, respectively. The increase of OprI hydrophobicity upon AMP binding revealed that it undergoes conformational changes for membrane fusion. Our results showed that OprI on bacterial surfaces is responsible for the recruitment and susceptibility to amphipathic α-helical AMPs and may be used to screen antimicrobials.
Collapse
|
48
|
Chu S, Kaur H, Nemati A, Walsh JD, Partida V, Zhang SQ, Gochin M. Swapped-domain constructs of the glycoprotein-41 ectodomain are potent inhibitors of HIV infection. ACS Chem Biol 2015; 10:1247-57. [PMID: 25646644 DOI: 10.1021/cb501021j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The conformational rearrangement of N- and C-heptad repeats (NHR, CHR) of the HIV-1 glycoprotein-41 (gp41) ectodomain into a trimer of hairpins triggers virus-cell fusion by bringing together membrane-spanning N- and C-terminal domains. Peptides derived from the NHR and CHR inhibit fusion by targeting a prehairpin intermediate state of gp41. Typically, peptides derived from the CHR are low nanomolar inhibitors, whereas peptides derived from the NHR are low micromolar inhibitors. Here, we describe the inhibitory activity of swapped-domain gp41 mimics of the form CHR-loop-NHR, which were designed to form reverse hairpin trimers exposing NHR grooves. We observed low nanomolar inhibition of HIV fusion in constructs that possessed the following properties: an extended NHR C-terminus, an exposed conserved hydrophobic pocket on the NHR, high helical content, and trimer stability. Low nanomolar activity was independent of CHR length. CD studies in membrane mimetic dodecylphosphocholine micelles suggested that bioactivity could be related to the ability of the inhibitors to interact with a membrane-associated prehairpin intermediate. The swapped-domain design resolves the problem of unstable and weakly active NHR peptides and suggests a different mechanism of action from that of CHR peptides in inhibition of HIV-1 fusion.
Collapse
Affiliation(s)
- Shidong Chu
- Department
of Basic Sciences, Touro University−California, Vallejo, California 94592, United States
| | - Hardeep Kaur
- Department
of Basic Sciences, Touro University−California, Vallejo, California 94592, United States
| | - Ariana Nemati
- Department
of Basic Sciences, Touro University−California, Vallejo, California 94592, United States
| | - Joseph D. Walsh
- Department
of Basic Sciences, Touro University−California, Vallejo, California 94592, United States
- Department
of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| | - Vivian Partida
- Department
of Basic Sciences, Touro University−California, Vallejo, California 94592, United States
| | - Shao-Qing Zhang
- Department
of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
- Department
of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19014, United States
| | - Miriam Gochin
- Department
of Basic Sciences, Touro University−California, Vallejo, California 94592, United States
- Department
of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| |
Collapse
|
49
|
Zhang Y, Bartz R, Grigoryan G, Bryant M, Aaronson J, Beck S, Innocent N, Klein L, Procopio W, Tucker T, Jadhav V, Tellers DM, DeGrado WF. Computational design and experimental characterization of peptides intended for pH-dependent membrane insertion and pore formation. ACS Chem Biol 2015; 10:1082-93. [PMID: 25630033 DOI: 10.1021/cb500759p] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
There are many opportunities to use macromolecules, such as peptides and oligonucleotides, for intracellular applications. Despite this, general methods for delivering these molecules to the cytosol in a safe and efficient manner are not available. Efforts to develop a variety of intracellular drug delivery systems such as viral vectors, lipoplexes, nanoparticles, and amphiphilic peptides have been made, but various challenges such as delivery efficiency, toxicity, and controllability remain. A central challenge is the ability to selectively perturb, not destroy, the membrane to facilitate cargo introduction. Herein, we describe our efforts to design and characterize peptides that form pores inside membranes at acidic pH, so-called pH-switchable pore formation (PSPF) peptides, as a potential means for facilitating cargo translocation through membranes. Consistent with pore formation, these peptides exhibit low-pH-triggered selective release of ATP and miRNA, but not hemoglobin, from red blood cells. Consistent with these observations, biophysical studies (tryptophan fluorescence, circular dichroism, size-exclusion chromatography, analytical ultracentrifugation, and attenuated total reflectance Fourier transformed infrared spectroscopy) show that decreased pH destabilizes the PSPF peptides in aqueous systems while promoting their membrane insertion. Together, these results suggest that reduced pH drives insertion of PSPF peptides into membranes, leading to target-specific escape through a proposed pore formation mechanism.
Collapse
Affiliation(s)
- Yao Zhang
- Department
of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - René Bartz
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Gevorg Grigoryan
- Department
of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Michael Bryant
- Department
of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jeff Aaronson
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Stephen Beck
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Nathalie Innocent
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Lee Klein
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - William Procopio
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Tom Tucker
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Vasant Jadhav
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - David M. Tellers
- Merck Research Laboratories, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - William F. DeGrado
- Department
of Pharmaceutical Chemistry, University of California−San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
50
|
Chong YC, Mann RK, Zhao C, Kato M, Beachy PA. Bifurcating action of Smoothened in Hedgehog signaling is mediated by Dlg5. Genes Dev 2015; 29:262-76. [PMID: 25644602 PMCID: PMC4318143 DOI: 10.1101/gad.252676.114] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 12/29/2014] [Indexed: 12/19/2022]
Abstract
Binding of the Hedgehog (Hh) protein signal to its receptor, Patched, induces accumulation of the seven-pass transmembrane protein Smoothened (Smo) within the primary cilium and of the zinc finger transcription factor Gli2 at the ciliary tip, resulting ultimately in Gli-mediated changes in nuclear gene expression. However, the mechanism by which pathway activation is communicated from Smo to Gli2 is not known. In an effort to elucidate this mechanism, we identified Dlg5 (Discs large, homolog 5) in a biochemical screen for proteins that preferentially interact with activated Smo. We found that disruption of Smo-Dlg5 interactions or depletion of endogenous Dlg5 leads to diminished Hh pathway response without a significant impact on Smo ciliary accumulation. We also found that Dlg5 is localized at the basal body, where it associates with another pathway component, Kif7. We show that Dlg5 is required for Hh-induced enrichment of Kif7 and Gli2 at the tip of the cilium but is dispensable for Gpr161 exit from the cilium and the consequent suppression of Gli3 processing into its repressor form. Our findings suggest a bifurcation of Smo activity in Hh response, with a Dlg5-independent arm for suppression of Gli repressor formation and a second arm involving Smo interaction with Dlg5 for Gli activation.
Collapse
Affiliation(s)
- Yong Chun Chong
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA; Department of Developmental Biology, Stanford University, Stanford, California 94305, USA; Department of Biochemistry, Stanford University, Stanford, California 94305, USA
| | - Randall K Mann
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA; Department of Developmental Biology, Stanford University, Stanford, California 94305, USA; Department of Biochemistry, Stanford University, Stanford, California 94305, USA
| | - Chen Zhao
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA; Department of Developmental Biology, Stanford University, Stanford, California 94305, USA; Department of Biochemistry, Stanford University, Stanford, California 94305, USA
| | - Masaki Kato
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA; Department of Developmental Biology, Stanford University, Stanford, California 94305, USA
| | - Philip A Beachy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA; Department of Developmental Biology, Stanford University, Stanford, California 94305, USA; Department of Biochemistry, Stanford University, Stanford, California 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
| |
Collapse
|