151
|
Blocquel D, Beltrandi M, Erales J, Barbier P, Longhi S. Biochemical and structural studies of the oligomerization domain of the Nipah virus phosphoprotein: evidence for an elongated coiled-coil homotrimer. Virology 2013; 446:162-72. [PMID: 24074578 DOI: 10.1016/j.virol.2013.07.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 07/08/2013] [Accepted: 07/24/2013] [Indexed: 12/19/2022]
Abstract
Nipah virus (NiV) is a recently emerged severe human pathogen that belongs to the Henipavirus genus within the Paramyxoviridae family. The NiV genome is encapsidated by the nucleoprotein (N) within a helical nucleocapsid that is the substrate used by the polymerase for transcription and replication. The polymerase is recruited onto the nucleocapsid via its cofactor, the phosphoprotein (P). The NiV P protein has a modular organization, with alternating disordered and ordered domains. Among these latter, is the P multimerization domain (PMD) that was predicted to adopt a coiled-coil conformation. Using both biochemical and biophysical approaches, we show that NiV PMD forms a highly stable and elongated coiled-coil trimer, a finding in striking contrast with respect to the PMDs of Paramyxoviridae members investigated so far that were all found to tetramerize. The present results therefore represent the first report of a paramyxoviral P protein forming trimers.
Collapse
Affiliation(s)
- David Blocquel
- CNRS and Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques (AFMB), UMR 7257, 13288 Marseille, France
| | | | | | | | | |
Collapse
|
152
|
Hamaguchi M, Kamikubo H, Suzuki KN, Hagihara Y, Yanagihara I, Sakata I, Kataoka M, Hamada D. Structural basis of α-catenin recognition by EspB from enterohaemorrhagic E. coli based on hybrid strategy using low-resolution structural and protein dissection. PLoS One 2013; 8:e71618. [PMID: 23967227 PMCID: PMC3743801 DOI: 10.1371/journal.pone.0071618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 07/02/2013] [Indexed: 01/29/2023] Open
Abstract
Enterohaemorrhagic E. coli (EHEC) induces actin reorganization of host cells by injecting various effectors into host cytosol through type III secretion systems. EspB is the natively partially folded EHEC effector which binds to host α-catenin to promote the actin bundling. However, its structural basis is poorly understood. Here, we characterize the overall structural properties of EspB based on low-resolution structural data in conjunction with protein dissection strategy. EspB showed a unique thermal response involving cold denaturation in the presence of denaturant according to far-UV circular dichroism (CD). Small angle X-ray scattering revealed the formation of a highly extended structure of EspB comparable to the ideal random coil. Various disorder predictions as well as CD spectra of EspB fragments identified the presence of α-helical structures around G41 to Q70. The fragment corresponding to this region indicated the thermal response similar to EspB. Moreover, this fragment showed a high affinity to C-terminal vinculin homology domain of α-catenin. The results clarified the importance of preformed α-helix of EspB for recognition of α-catenin.
Collapse
Affiliation(s)
- Mitsuhide Hamaguchi
- Department of Emergency Critical Care Medicine, School of Medicine, Kinki University, Osakasayama, Osaka, Japan
- Research Institute, Osaka Medical Center for Maternal and Child Health, Izumi, Japan
| | - Hironari Kamikubo
- Laboratory of Bioenergetics and Biophysics, Nara Institute of Science and Technology (NAIST), Ikoma, Nara, Japan
| | - Kayo N. Suzuki
- Research Institute, Osaka Medical Center for Maternal and Child Health, Izumi, Japan
| | - Yoshihisa Hagihara
- National Institute of Advanced Industrial Science and Technology (AIST), Ikeda, Osaka, Japan
| | - Itaru Yanagihara
- Research Institute, Osaka Medical Center for Maternal and Child Health, Izumi, Japan
| | - Ikuhiro Sakata
- Department of Emergency Critical Care Medicine, School of Medicine, Kinki University, Osakasayama, Osaka, Japan
| | - Mikio Kataoka
- Laboratory of Bioenergetics and Biophysics, Nara Institute of Science and Technology (NAIST), Ikoma, Nara, Japan
| | - Daizo Hamada
- Division of Structural Biology, Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Kobe University, Chuo-ku, Kobe, Japan
| |
Collapse
|
153
|
Quistgaard EM, Löw C, Moberg P, Guettou F, Maddi K, Nordlund P. Structural and biophysical characterization of the cytoplasmic domains of human BAP29 and BAP31. PLoS One 2013; 8:e71111. [PMID: 23967155 PMCID: PMC3742741 DOI: 10.1371/journal.pone.0071111] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 06/24/2013] [Indexed: 11/18/2022] Open
Abstract
Two members of the B-cell associated 31 (BAP31) family are found in humans; BAP29 and BAP31. These are ubiquitously expressed receptors residing in the endoplasmic reticulum. BAP31 functions in sorting of membrane proteins and in caspase-8 mediated apoptosis, while BAP29 appears to mainly corroborate with BAP31 in sorting. The N-terminal half of these proteins is membrane-bound while the C-terminal half is cytoplasmic. The latter include the so called variant of death effector domain (vDED), which shares weak sequence homology with DED domains. Here we present two structures of BAP31 vDED determined from a single and a twinned crystal, grown at pH 8.0 and pH 4.2, respectively. These structures show that BAP31 vDED forms a dimeric parallel coiled coil with no structural similarity to DED domains. Solution studies support this conclusion and strongly suggest that an additional α-helical domain is present in the C-terminal cytoplasmic region, probably forming a second coiled coil. The thermal stability of BAP31 vDED is quite modest at neutral pH, suggesting that it may assemble in a dynamic fashion in vivo. Surprisingly, BAP29 vDED is partially unfolded at pH 7, while a coiled coil is formed at pH 4.2 in vitro. It is however likely that folding of the domain is triggered by other factors than low pH in vivo. We found no evidence for direct interaction of the cytoplasmic domains of BAP29 and BAP31.
Collapse
Affiliation(s)
- Esben M. Quistgaard
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (EMQ); (PN)
| | - Christian Löw
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Per Moberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Fatma Guettou
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Karthik Maddi
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Pär Nordlund
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- * E-mail: (EMQ); (PN)
| |
Collapse
|
154
|
Ros U, Souto ALCF, de Oliveira FJ, Crusca E, Pazos F, Cilli EM, Lanio ME, Schreier S, Alvarez C. Functional and topological studies with Trp-containing analogs of the peptide StII1-30derived from the N-terminus of the pore forming toxin sticholysin II: contribution to understand its orientation in membrane. Biopolymers 2013; 100:337-46. [DOI: 10.1002/bip.22211] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 12/14/2012] [Accepted: 01/14/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Uris Ros
- Center for Protein Studies; Biology Faculty; University of Havana (UH); Havana; Cuba
| | - Ana Lucia C. F. Souto
- Department of Biochemistry; Institute of Chemistry; University of São Paulo (USP); São Paulo; Brazil
| | - Felipe J. de Oliveira
- Department of Biochemistry; Institute of Chemistry; University of São Paulo (USP); São Paulo; Brazil
| | - Edson Crusca
- Department of Biochemistry and Chemical Technology; Institute of Chemistry; São Paulo State University (UNESP); Araraquara; São Paulo; Brazil
| | - Fabiola Pazos
- Center for Protein Studies; Biology Faculty; University of Havana (UH); Havana; Cuba
| | - Eduardo M. Cilli
- Department of Biochemistry and Chemical Technology; Institute of Chemistry; São Paulo State University (UNESP); Araraquara; São Paulo; Brazil
| | - Maria E. Lanio
- Center for Protein Studies; Biology Faculty; University of Havana (UH); Havana; Cuba
| | - Shirley Schreier
- Department of Biochemistry; Institute of Chemistry; University of São Paulo (USP); São Paulo; Brazil
| | - Carlos Alvarez
- Center for Protein Studies; Biology Faculty; University of Havana (UH); Havana; Cuba
| |
Collapse
|
155
|
Bhattacharjee P, Banerjee A, Banerjee A, Dasgupta D, Sengupta K. Structural Alterations of Lamin A Protein in Dilated Cardiomyopathy. Biochemistry 2013; 52:4229-41. [DOI: 10.1021/bi400337t] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Pritha Bhattacharjee
- Biophysics
Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Avinanda Banerjee
- Biophysics
Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Amrita Banerjee
- Biophysics
Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Dipak Dasgupta
- Biophysics
Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Kaushik Sengupta
- Biophysics
Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| |
Collapse
|
156
|
Malik L, Nygaard J, Christensen NJ, Streicher WW, Thulstrup PW, Arleth L, Jensen KJ. Self-assembly of designed coiled coil peptides studied by small-angle X-ray scattering and analytical ultracentrifugation. J Pept Sci 2013; 19:283-92. [DOI: 10.1002/psc.2497] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 01/23/2013] [Accepted: 01/24/2013] [Indexed: 11/08/2022]
Affiliation(s)
- Leila Malik
- University of Copenhagen; Thorvaldsensvej 40 DK-1871 Frederiksberg C Denmark
| | - Jesper Nygaard
- University of Copenhagen; Thorvaldsensvej 40 DK-1871 Frederiksberg C Denmark
- University of Lund; Ole Römers väg 1 SE-223 63 Lund Sweden
| | | | - Werner W. Streicher
- Faculty of Health Science; NNF Center for Protein Research; Blegdamsvej 3B DK-2200 Copenhagen N Denmark
| | - Peter W. Thulstrup
- University of Copenhagen; Thorvaldsensvej 40 DK-1871 Frederiksberg C Denmark
| | - Lise Arleth
- University of Copenhagen; Thorvaldsensvej 40 DK-1871 Frederiksberg C Denmark
| | - Knud J. Jensen
- University of Copenhagen; Thorvaldsensvej 40 DK-1871 Frederiksberg C Denmark
| |
Collapse
|
157
|
Zhang S, Li Z. Stimuli-responsive polypeptide materials prepared by ring-opening polymerization of α-amino acidN-carboxyanhydrides. ACTA ACUST UNITED AC 2013. [DOI: 10.1002/polb.23263] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
158
|
The α-helical regions of KERP1 are important in Entamoeba histolytica adherence to human cells. Sci Rep 2013; 3:1171. [PMID: 23378906 PMCID: PMC3558696 DOI: 10.1038/srep01171] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 12/27/2012] [Indexed: 11/19/2022] Open
Abstract
The lysine and glutamic acid rich protein KERP1 is a unique surface adhesion factor associated with virulence in the human pathogen Entamoeba histolytica. Both the function and structure of this protein remain unknown to this date. Here, we used circular dichroism, analytical ultracentrifugation and bioinformatics modeling to characterize the structure of KERP1. Our findings revealed that it is an α-helical rich protein organized as a trimer, endowed with a very high thermal stability (Tm = 89.6°C). Bioinformatics sequence analyses and 3D-structural modeling indicates that KERP1 central segments could account for protein trimerization. Relevantly, expressing the central region of KERP1 in living parasites, impair their capacity to adhere to human cells. Our observations suggest a link between the inhibitory effect of the isolated central region and the structural features of KERP1.
Collapse
|
159
|
Zhang S, Chen C, Li Z. Effects of molecular weight on thermal responsive property of pegylated poly-l-glutamates. CHINESE JOURNAL OF POLYMER SCIENCE 2013. [DOI: 10.1007/s10118-013-1218-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
160
|
DiMarco RL, Heilshorn SC. Multifunctional materials through modular protein engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2012; 24:3923-40. [PMID: 22730248 DOI: 10.1002/adma.201200051] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Indexed: 05/20/2023]
Abstract
The diversity of potential applications for protein-engineered materials has undergone profound recent expansion through a rapid increase in the library of domains that have been utilized in these materials. Historically, protein-engineered biomaterials have been generated from a handful of peptides that were selected and exploited for their naturally evolved functionalities. In recent years, the scope of the field has drastically expanded to include peptide domains that were designed through computational modeling, identified through high-throughput screening, or repurposed from wild type domains to perform functions distinct from their primary native applications. The strategy of exploiting a diverse library of peptide domains to design modular block copolymers enables the synthesis of multifunctional protein-engineered materials with a range of customizable properties and activities. As the diversity of peptide domains utilized in modular protein engineering continues to expand, a tremendous and ever-growing combinatorial expanse of material functionalities will result.
Collapse
|
161
|
Bains GK, Kim SH, Sorin EJ, Narayanaswami V. The extent of pyrene excimer fluorescence emission is a reflector of distance and flexibility: analysis of the segment linking the LDL receptor-binding and tetramerization domains of apolipoprotein E3. Biochemistry 2012; 51:6207-19. [PMID: 22779734 DOI: 10.1021/bi3005285] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Pyrene is a spatially sensitive probe that displays an ensemble of monomeric fluorescence emission peaks (375-405 nm) and an additional band (called excimer) at ~460 nm when two fluorophores are spatially proximal. We examined if there is a correlation between distance between two pyrenes on an α-helical structure and excimer/monomer (e/m) ratio. Using structure-guided design, pyrene maleimide was attached to pairs of Cys residues separated by ~5 Å increments on helix 2 of the N-terminal domain of apolipoprotein E3 (apoE3). Fluorescence spectral analysis revealed an intense excimer band when the probes were ~5 Å from each other with an e/m ratio of ~3.0, which decreased to ~1.0 at 20 Å. An inverse correlation between e/m ratio and the distance between pyrenes was observed, with the probe and helix flexibility also contributing to the extent of excimer formation. We verified this approach by estimating the distance between T57C and C112 (located on helices 2 and 3, respectively) to be 5.2 Å (4.9 Å from NMR and 5.7 Å from the X-ray structure). Excimer formation was also noted to a significant extent with probes located in the linker segment, suggesting spatial proximity (10-15 Å) to corresponding sites on neighboring molecules in the tetrameric configuration of apoE. We infer that oligomerization via the C-terminal domain juxtaposes the linker segments from neighboring apoE molecules. This study offers new insights into the conformation of tetrameric apoE and presents the use of pyrene as a powerful probe for studying protein spatial organization.
Collapse
Affiliation(s)
- Gursharan K Bains
- Department of Chemistry and Biochemistry, 1250 Bellflower Boulevard, California State University Long Beach, Long Beach, CA 90840, USA
| | | | | | | |
Collapse
|
162
|
Morstadt L, Meng QC, Johansson JS. Design and biophysical characterization of a monomeric four-alpha-helix bundle protein Aα₄ with affinity for the volatile anesthetic halothane. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2012; 1824:1409-15. [PMID: 22750405 DOI: 10.1016/j.bbapap.2012.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 06/06/2012] [Accepted: 06/14/2012] [Indexed: 11/15/2022]
Abstract
A monomeric four-α-helix bundle protein Aα₄ was designed as a step towards investigating the interaction of volatile general anesthetics with their putative membrane protein targets. The alpha helices, connected by glycine loops, have the sequence A, B, B', A'. The DNA sequence was designed to make the helices with the same amino acid sequences (helix A and A', B and B', respectively) as different as possible, while using codons which are favorable for expression in E. coli. The protein was bacterially expressed and purified to homogeneity using reversed-phase HPLC. Protein identity was verified using MALDI-TOF mass spectrometry. Far-UV circular dichroism spectroscopy confirmed the predominantly alpha-helical nature of the protein Aα₄. Guanidinium chloride induced denaturation showed that the monomeric four-α-helix bundle protein Aα₄ is considerably more stable compared to the dimeric di-α-helical protein (Aα₂-L38M)₂. The sigmoidal character of the unfolding reaction is conserved while the sharpness of the transition is increased 1.8-fold. The monomeric four-α-helix bundle protein Aα₄ bound halothane with a dissociation constant (K(d)) of 0.93 ± 0.02mM, as shown by both tryptophan fluorescence quenching and isothermal titration calorimetry. This monomeric four-α-helix bundle protein can now be used as a scaffold to incorporate natural central nervous system membrane protein sequences in order to examine general anesthetic interactions with putative targets in detail.
Collapse
Affiliation(s)
- Lucia Morstadt
- University of Pennsylvania School of Medicine, Department of Anesthesiology and Critical Care, 3620 Hamilton Walk, Philadelphia, PA 19104, USA.
| | | | | |
Collapse
|
163
|
Nevzorov IA, Levitsky DI. Tropomyosin: double helix from the protein world. BIOCHEMISTRY (MOSCOW) 2012; 76:1507-27. [PMID: 22339601 DOI: 10.1134/s0006297911130098] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This review concerns the structure and functions of tropomyosin (TM), an actin-binding protein that plays a key role in the regulation of muscle contraction. The TM molecule is a dimer of α-helices, which form a coiled-coil. Recent views on the TM structure are analyzed, and special attention is concentrated on those structural traits of the TM molecule that distinguish it from the other coiled-coil proteins. Modern data are presented on TM functional properties, such as its interaction with actin and ability to move on the surface of actin filaments, which underlies the regulation of the actin-myosin interaction upon contraction of skeletal and cardiac muscles. Also, part of the review is devoted to analysis of the effects of mutations in TM genes associated with muscle diseases (myopathies) on the structure and functions of TM.
Collapse
Affiliation(s)
- I A Nevzorov
- Bach Institute of Biochemistry, Russian Academy of Sciences, Moscow, Russia.
| | | |
Collapse
|
164
|
Harrison JS, Koellhoffer JF, Chandran K, Lai JR. Marburg virus glycoprotein GP2: pH-dependent stability of the ectodomain α-helical bundle. Biochemistry 2012; 51:2515-25. [PMID: 22369502 DOI: 10.1021/bi3000353] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Marburg virus (MARV) and Ebola virus (EBOV) constitute the family Filoviridae of enveloped viruses (filoviruses) that cause severe hemorrhagic fever. Infection by MARV requires fusion between the host cell and viral membranes, a process that is mediated by the two subunits of the envelope glycoprotein, GP1 (surface subunit) and GP2 (transmembrane subunit). Upon viral attachment and uptake, it is believed that the MARV viral fusion machinery is triggered by host factors and environmental conditions found in the endosome. Next, conformational rearrangements in the GP2 ectodomain result in the formation of a highly stable six-helix bundle; this refolding event provides the energetic driving force for membrane fusion. Both GP1 and GP2 from EBOV have been extensively studied, but there is little information available for the MARV glycoproteins. Here we have expressed two variants of the MARV GP2 ectodomain in Escherichia coli and analyzed their biophysical properties. Circular dichroism indicates that the MARV GP2 ectodomain adopts an α-helical conformation, and one variant sediments as a trimer by equilibrium analytical ultracentrifugation. Denaturation studies indicate the α-helical structure is highly stable at pH 5.3 (unfolding energy, ΔG(unf,H(2)O), of 33.4 ± 2.5 kcal/mol and melting temperature, T(m), of 75.3 ± 2.1 °C for one variant). Furthermore, we found the α-helical stability to be strongly dependent on pH, with higher stability under lower-pH conditions (T(m) values ranging from ~92 °C at pH 4.0 to ~38 °C at pH 8.0). Mutational analysis suggests two glutamic acid residues (E579 and E580) are partially responsible for this pH-dependent behavior. On the basis of these results, we hypothesize that the pH-dependent folding stability of the MARV GP2 ectodomain provides a mechanism for controlling conformational preferences such that the six-helix bundle "postfusion" state is preferred under conditions of appropriately matured endosomes.
Collapse
Affiliation(s)
- Joseph S Harrison
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | | | | | | |
Collapse
|
165
|
Tessera V, Guida F, Juretić D, Tossi A. Identification of antimicrobial peptides from teleosts and anurans in expressed sequence tag databases using conserved signal sequences. FEBS J 2012; 279:724-36. [PMID: 22188679 DOI: 10.1111/j.1742-4658.2011.08463.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The problem of multidrug resistance requires the efficient and accurate identification of new classes of antimicrobial agents. Endogenous antimicrobial peptides produced by most organisms are a promising source of such molecules. We have exploited the high conservation of signal sequences in teleost and anuran antimicrobial peptides to search cDNA (expressed sequence tag) databases for likely candidates. Subject sequences were then analysed for the presence of potential antimicrobial peptides based on physicochemical properties (amphipathic helical structure, cationicity) and use of the D-descriptor model to predict the therapeutic index (relation between the minimum inhibitory concentration and the concentration giving 50% haemolysis). This analysis also suggested mutations to probe the role of the primary structure in determining potency and selectivity. Selected sequences were chemically synthesized and the antimicrobial activity of the peptides was confirmed. In particular, a short (21-residue) sequence, likely of sticklefish origin, showed potent activity and it was possible to tune the spectrum of action and/or selectivity by combining three directed mutations. Membrane permeabilization studies on both bacterial and host cells indicate that the mode of action was prevalently membranolytic. This method opens up the possibility for more effective searching of the vast and continuously growing expressed sequence tag databases for novel antimicrobial peptides, which are likely abundant, and the efficient identification of the most promising candidates among them.
Collapse
|
166
|
Size-exclusion chromatography in structural analysis of intrinsically disordered proteins. Methods Mol Biol 2012; 896:179-94. [PMID: 22821524 DOI: 10.1007/978-1-4614-3704-8_11] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Gel-filtration chromatography, also known as size-exclusion chromatography (SEC) or gel-permeation chromatography, is a useful tool for structural and conformational analyses of intrinsically disordered proteins (IDPs). SEC can be utilized for the estimation of the hydrodynamic dimensions of a given IDP, for evaluation of the association state, for the analysis of IDP interactions with binding partners, and for the induced folding studies. It also can be used to physically separate IDP conformers based on their hydrodynamic dimensions, thus providing a unique possibility for the independent analysis of their physicochemical properties.
Collapse
|
167
|
Solmaz SR, Chauhan R, Blobel G, Melčák I. Molecular architecture of the transport channel of the nuclear pore complex. Cell 2011; 147:590-602. [PMID: 22036567 PMCID: PMC3431207 DOI: 10.1016/j.cell.2011.09.034] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2011] [Revised: 06/28/2011] [Accepted: 09/12/2011] [Indexed: 01/02/2023]
Abstract
The nuclear pore complex encloses a central channel for nucleocytoplasmic transport, which is thought to consist of three nucleoporins, Nup54, Nup58, and Nup62. However, the structure and composition of the channel are elusive. We determined the crystal structures of the interacting domains between these nucleoporins and pieced together the molecular architecture of the mammalian transport channel. Located in the channel midplane is a flexible Nup54⋅Nup58 ring that can undergo large rearrangements yielding diameter changes from ∼20 to ∼40 nm. Nup62⋅Nup54 triple helices project alternately up and down from either side of the midplane ring and form nucleoplasmic and cytoplasmic entries. The channel consists of as many as 224 copies of the three nucleoporins, amounting to a molar mass of 12.3 MDa and contributing 256 phenylalanine-glycine repeat regions. We propose that the occupancy of these repeat regions with transport receptors modulates ring diameter and transport activity.
Collapse
Affiliation(s)
- Sozanne R Solmaz
- Laboratory of Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | | | | | | |
Collapse
|
168
|
Interaction between sodium dodecyl sulfate and membrane reconstituted aquaporins: A comparative study of spinach SoPIP2;1 and E. coli AqpZ. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1808:2600-7. [DOI: 10.1016/j.bbamem.2011.05.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 05/27/2011] [Accepted: 05/31/2011] [Indexed: 01/13/2023]
|
169
|
Li GR, He LY, Liu XY, Liu AP, Huang YB, Qiu C, Zhang XY, Xu JQ, Yang W, Chen YX. Rational design of peptides with anti-HCV/HIV activities and enhanced specificity. Chem Biol Drug Des 2011; 78:835-43. [PMID: 21801309 DOI: 10.1111/j.1747-0285.2011.01201.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Lack of vaccines for HCV and HIV makes the antiviral drug development urgently needed. The recently identified HCV NS5A-derived virucidal peptide (C5A) demonstrated a wide spectrum of activities against viruses. In this study, the C5A sequence SWLRDIWDWICEVLSDFK was utilized as the framework to study the effect of the modulation of peptide helicity and hydrophobicity on its anti-HCV and anti-HIV activities. Peptide helicity and hydrophobicity were altered by substitutions of varying amino acids on the non-polar face of C5A. Peptide hydrophobicity has been proved to play a crucial role in peptide anti-HCV or anti-HIV activities. Peptide helicity was relatively independent with antiviral activity. However, peptide analogs with dimerized structure in an aqueous medium while maintaining the ability to be induced into a more helical structure in a hydrophobic environment may tend to show comparable or improved antiviral activity and specificity to C5A. By modulating peptide helicity and hydrophobicity, we improved the specificity of C5A against HCV and HIV by 23- and 69-fold, respectively, in terms of the ratio of hemolytic activity to antiviral activity. We demonstrated that obtained by de novo design approach, peptide I6L/I10L/V13L may be a promising candidate as a new anti-HCV and anti-HIV therapeutic.
Collapse
Affiliation(s)
- Gui-Rong Li
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, Changchun, Jilin, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Crooks RO, Rao T, Mason JM. Truncation, randomization, and selection: generation of a reduced length c-Jun antagonist that retains high interaction stability. J Biol Chem 2011; 286:29470-9. [PMID: 21697091 PMCID: PMC3190987 DOI: 10.1074/jbc.m111.221267] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 05/17/2011] [Indexed: 12/22/2022] Open
Abstract
The DNA binding activity of the transcriptional regulator activator protein-1 shows considerable promise as a target in cancer therapy. A number of different strategies have been employed to inhibit the function of this protein with promise having been demonstrated both in vitro and in vivo. Peptide-based therapeutics have received renewed interest in the last few years, and a number of 37-amino acid peptides capable of binding to the coiled coil dimerization domain of Jun and Fos have been derived. Here, we demonstrate how truncation and semi-rational library design, followed by protein-fragment complementation, can be used to produce a leucine zipper binding peptide by iterative means. To this end, we have implemented this strategy on the FosW peptide to produce 4hFosW. This peptide is truncated by four residues with comparably favorable binding properties and demonstrates the possibility to design progressively shorter peptides to serve as leucine zipper antagonists while retaining many of the key features of the parent peptide. Whether or not the necessity for low molecular weight antagonists is required from the perspective of druggability and efficacy is subject to debate. However, antagonists of reduced length are worthy of perusal from the point of view of synthetic cost as well as identifying the smallest functional unit that is required for binding.
Collapse
Affiliation(s)
- Richard O. Crooks
- From the Department of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, United Kingdom
| | - Tara Rao
- From the Department of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, United Kingdom
| | - Jody M. Mason
- From the Department of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, United Kingdom
| |
Collapse
|
171
|
SCM, a novel M-like protein from Streptococcus canis, binds (mini)-plasminogen with high affinity and facilitates bacterial transmigration. Biochem J 2011; 434:523-35. [PMID: 21210764 DOI: 10.1042/bj20101121] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Streptococcus canis is an important zoonotic pathogen capable of causing serious invasive diseases in domestic animals and humans. In the present paper we report the binding of human plasminogen to S. canis and the recruitment of proteolytically active plasmin on its surface. The binding receptor for plasminogen was identified as a novel M-like protein designated SCM (S. canis M-like protein). SPR (surface plasmon resonance) analyses, radioactive dot-blot analyses and heterologous expression on the surface of Streptococcus gordonii confirmed the plasminogen-binding capability of SCM. The binding domain was located within the N-terminus of SCM, which specifically bound to the C-terminal part of plasminogen (mini-plasminogen) comprising kringle domain 5 and the catalytic domain. In the presence of urokinase, SCM mediated plasminogen activation on the bacterial surface that was inhibited by serine protease inhibitors and lysine amino acid analogues. Surface-bound plasmin effectively degraded purified fibrinogen as well as fibrin clots, resulting in the dissolution of fibrin thrombi. Electron microscopic illustration and time-lapse imaging demonstrated bacterial transmigration through fibrinous thrombi. The present study has led, for the first time, to the identification of SCM as a novel receptor for (mini)-plasminogen mediating the fibrinolytic activity of S. canis.
Collapse
|
172
|
Abstract
Expression of a retroviral protein, Gag, in mammalian cells is sufficient for assembly of immature virus-like particles (VLPs). VLP assembly is mediated largely by interactions between the capsid (CA) domains of Gag molecules but is facilitated by binding of the nucleocapsid (NC) domain to nucleic acid. We have investigated the role of SP1, a spacer between CA and NC in HIV-1 Gag, in VLP assembly. Mutational analysis showed that even subtle changes in the first 4 residues of SP1 destroy the ability of Gag to assemble correctly, frequently leading to formation of tubes or other misassembled structures rather than proper VLPs. We also studied the conformation of the CA-SP1 junction region in solution, using both molecular dynamics simulations and circular dichroism. Consonant with nuclear magnetic resonance (NMR) studies from other laboratories, we found that SP1 is nearly unstructured in aqueous solution but undergoes a concerted change to an α-helical conformation when the polarity of the environment is reduced by addition of dimethyl sulfoxide (DMSO), trifluoroethanol, or ethanol. Remarkably, such a coil-to-helix transition is also recapitulated in an aqueous medium at high peptide concentrations. The exquisite sensitivity of SP1 to mutational changes and its ability to undergo a concentration-dependent structural transition raise the possibility that SP1 could act as a molecular switch to prime HIV-1 Gag for VLP assembly. We suggest that changes in the local environment of SP1 when Gag oligomerizes on nucleic acid might trigger this switch.
Collapse
|
173
|
Kashiwada A, Tsuboi M, Matsuda K. Target-selective one-way membrane fusion system based on a pH-responsive coiled coil assembly at the interface of liposomal vesicles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2011; 27:1403-1408. [PMID: 21058731 DOI: 10.1021/la103908u] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The coiled coil trimer structure is a common motif observed in membrane fusion processes of specific fusion proteins such as the hemagglutinin glycoprotein. The HA2 subunit in the hemagglutinin changes its conformation or geometry to be favorable to membrane fusion in response to endosomal weakly acidic pH. This pH responsiveness is indispensable to an artificial polypeptide-triggered delivery system as well as the membrane fusion reaction in biology. In this study, we have constructed an AAB-type coiled coil heteroassembled system that is sensitive to weakly acidic pH. The heterotrimer is formed from two kinds of polypeptides containing an Ala or a Trp residue at a hydrophobic a position, and it was observed that the Glu residue at the other a position induced an acidic pH-dependent conformational change. On the basis of this pH-responsive coiled coil heteroassembled system, a boronic acid coupled working polypeptide for the combination of an intervesicular complex with a sugarlike compound on the surface of the target liposome, and a supporting polypeptide for the construction of a pH-responsive heterotrimer with the working polypeptide were designed and synthesized. The process of membrane fusion was characterized by lipid-mixing, inner-leaflet lipid-mixing, and content-mixing assays. The target selective vesicle fusion is clearly observed at a weakly acidic pH, where the working polypeptides form a heterotrimeric coiled coil with the supporting polypeptides in a 1:2 binding stoichiometry and the surfaces between pilot and target vesicles come into close proximity to each other.
Collapse
Affiliation(s)
- Ayumi Kashiwada
- Department of Applied Molecular Chemistry, Graduate School of Industrial Technology, Nihon University, Narashino, Chiba, Japan.
| | | | | |
Collapse
|
174
|
Abstract
Dimerization of the Jun-Fos activator protein-1 (AP-1) transcriptional regulator is mediated by coiled coil regions that facilitate binding of the basic regions to a specific promoter. AP-1 is responsible for the regulation of a number of genes involved in cell proliferation. We have previously derived peptide antagonists and demonstrated them to be capable of binding to the Jun or Fos coiled coil region with high affinity (K(D) values in the low nM range relative to μM for the wild-type interaction). Use of isothermal titration calorimetry combined with CD spectroscopy is reported to elucidate the thermodynamic parameters that drive the interaction stability of peptide antagonists with their cJun and cFos targets. We observe that the free energy of binding for antagonist-target complexes is dominated by the enthalpic term, is opposed by unfavourable entropic contributions consistent with reduced conformational freedom and that these values in turn correlate well (r = -0.97) with the measured helicity of each dimeric pair. The more helical the antagonist-target complex, the more favourable the change in enthalpy, which is in turn opposed more strongly by entropy. Antagonistic peptides are predicted to represent excellent scaffolds for further refinement. By contrast, the wild-type cJun-cFos complex is dominated by a favourable entropic contribution, owing partially to a decrease in buried hydrophobic groups from cFos core residues and an increase in the conformational freedom.
Collapse
|
175
|
Mokhtarzada S, Yu C, Brickenden A, Choy WY. Structural characterization of partially disordered human Chibby: insights into its function in the Wnt-signaling pathway. Biochemistry 2011; 50:715-26. [PMID: 21182262 PMCID: PMC3031990 DOI: 10.1021/bi101236z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
![]()
The Wnt/β-catenin signaling pathway is critical to embryonic development as well as adult tissue regeneration. Dysregulation of this pathway can lead to a variety of human diseases, in particular cancers. Chibby (Cby), a small and highly conserved protein, plays an antagonistic role in Wnt signaling by inhibiting the binding of β-catenin to Tcf/Lef family proteins, a protein interaction that is essential for the transcriptional activation of Wnt target genes. Cby is also involved in regulating intracellular distribution of β-catenin. Phosphorylated Cby forms a ternary complex with 14-3-3 protein and β-catenin, facilitating the export of β-catenin from the nucleus. On the other hand, the antagonistic function of Cby is inhibited upon binding to thyroid cancer-1 (TC-1). To dissect the structure−function relationship of Cby, we have used NMR spectroscopy, ESI-MS, CD, and DLS to extensively characterize the structure of human Cby. Our results show that the 126-residue Cby is partially disordered under nondenaturing conditions. While the N-terminal portion of the protein is predominantly unstructured in solution, the C-terminal half of Cby adopts a coiled-coil structure through self-association. Initial data for the binding studies of Cby to 14-3-3ζ (one of the isoforms in the 14-3-3 family) and TC-1 via these two distinct structural modules have also been obtained. It is noteworthy that in a recent large-scale analysis of the intrinsically disordered proteome of mouse, a substantial number of disordered proteins are predicted to have coiled-coil motif presence in their sequences. The combination of these two molecular recognition features could facilitate disordered Cby in assembling protein complexes via different modes of interaction.
Collapse
Affiliation(s)
- Sulayman Mokhtarzada
- Department of Biochemistry, The University of Western Ontario, London, Ontario, Canada N6A 5C1
| | | | | | | |
Collapse
|
176
|
King WJ, Murphy WL. Bioinspired conformational changes: an adaptable mechanism for bio-responsive protein delivery. Polym Chem 2011. [DOI: 10.1039/c0py00244e] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
177
|
Gradišar H, Jerala R. De novo design of orthogonal peptide pairs forming parallel coiled-coil heterodimers. J Pept Sci 2010; 17:100-6. [PMID: 21234981 DOI: 10.1002/psc.1331] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Revised: 10/20/2010] [Accepted: 10/20/2010] [Indexed: 11/07/2022]
Abstract
We used the principles governing the selectivity and stability of coiled-coil segments to design and experimentally test a set of four pairs of parallel coiled-coil-forming peptides composed of four heptad repeats. The design was based on maximizing the difference in stability between desired pairs and the most stable unwanted combinations using N-terminal helix initiator residues, favorable combinations of the electrostatic and hydrophobic interaction motifs and negative design motif based on burial of asparagine residues. Experimental analysis of all 36 pair combinations among the eight peptides was performed by circular dichroism (CD). On the basis of CD spectra, each peptide formed a high level of α-helical structure exclusively in combination with its designed peptide partner which demonstrates the orthogonality of the designed peptide pair set.
Collapse
Affiliation(s)
- Helena Gradišar
- Department of Biotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | | |
Collapse
|
178
|
A biophysical approach to phospholipase A2 activity and inhibition by anti-inflammatory drugs. Biophys Chem 2010; 152:109-17. [DOI: 10.1016/j.bpc.2010.08.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2010] [Revised: 08/14/2010] [Accepted: 08/17/2010] [Indexed: 11/18/2022]
|
179
|
Haque ME, Elmore KB, Tripathy A, Koc H, Koc EC, Spremulli LL. Properties of the C-terminal tail of human mitochondrial inner membrane protein Oxa1L and its interactions with mammalian mitochondrial ribosomes. J Biol Chem 2010; 285:28353-62. [PMID: 20601428 DOI: 10.1074/jbc.m110.148262] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In humans the mitochondrial inner membrane protein Oxa1L is involved in the biogenesis of membrane proteins and facilitates the insertion of both mitochondrial- and nuclear-encoded proteins from the mitochondrial matrix into the inner membrane. The C-terminal approximately 100-amino acid tail of Oxa1L (Oxa1L-CTT) binds to mitochondrial ribosomes and plays a role in the co-translational insertion of mitochondria-synthesized proteins into the inner membrane. Contrary to suggestions made for yeast Oxa1p, our results indicate that the C-terminal tail of human Oxa1L does not form a coiled-coil helical structure in solution. The Oxa1L-CTT exists primarily as a monomer in solution but forms dimers and tetramers at high salt concentrations. The binding of Oxa1L-CTT to mitochondrial ribosomes is an enthalpy-driven process with a K(d) of 0.3-0.8 microM and a stoichiometry of 2. Oxa1L-CTT cross-links to mammalian mitochondrial homologs of the bacterial ribosomal proteins L13, L20, and L28 and to mammalian mitochondrial specific ribosomal proteins MRPL48, MRPL49, and MRPL51. Oxa1L-CTT does not cross-link to proteins decorating the conventional exit tunnel of the bacterial large ribosomal subunit (L22, L23, L24, and L29).
Collapse
Affiliation(s)
- Md Emdadul Haque
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599-3290, USA
| | | | | | | | | | | |
Collapse
|
180
|
Oot RA, Wilkens S. Domain characterization and interaction of the yeast vacuolar ATPase subunit C with the peripheral stator stalk subunits E and G. J Biol Chem 2010; 285:24654-64. [PMID: 20529855 DOI: 10.1074/jbc.m110.136960] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The proton pumping activity of the eukaryotic vacuolar ATPase (V-ATPase) is regulated by a unique mechanism that involves reversible enzyme dissociation. In yeast, under conditions of nutrient depletion, the soluble catalytic V(1) sector disengages from the membrane integral V(o), and at the same time, both functional units are silenced. Notably, during enzyme dissociation, a single V(1) subunit, C, is released into the cytosol. The affinities of the other V(1) and V(o) subunits for subunit C are therefore of particular interest. The C subunit crystal structure shows that the subunit is elongated and dumbbell-shaped with two globular domains (C(head) and C(foot)) separated by a flexible helical neck region (Drory, O., Frolow, F., and Nelson, N. (2004) EMBO Rep. 5, 1148-1152). We have recently shown that subunit C is bound in the V(1)-V(o) interface where the subunit is in contact with two of the three peripheral stators (subunit EG heterodimers): one via C(head) and one via C(foot) (Zhang, Z., Zheng, Y., Mazon, H., Milgrom, E., Kitagawa, N., Kish-Trier, E., Heck, A. J., Kane, P. M., and Wilkens, S. (2008) J. Biol. Chem. 283, 35983-35995). In vitro, however, subunit C binds only one EG heterodimer (Féthière, J., Venzke, D., Madden, D. R., and Böttcher, B. (2005) Biochemistry 44, 15906-15914), implying that EG has different affinities for the two domains of the C subunit. To determine which subunit C domain binds EG with high affinity, we have generated C(head) and C(foot) and characterized their interaction with subunit EG heterodimer. Our findings indicate that the high affinity site for EGC interaction is C(head). In addition, we provide evidence that the EGC(head) interaction greatly stabilizes EG heterodimer.
Collapse
Affiliation(s)
- Rebecca A Oot
- Department of Biochemistry and Molecular Biology, State University of New York Upstate Medical University, Syracuse, New York 13210, USA
| | | |
Collapse
|
181
|
Eiríksdóttir E, Konate K, Langel Ü, Divita G, Deshayes S. Secondary structure of cell-penetrating peptides controls membrane interaction and insertion. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2010; 1798:1119-28. [DOI: 10.1016/j.bbamem.2010.03.005] [Citation(s) in RCA: 250] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 01/19/2010] [Accepted: 03/02/2010] [Indexed: 01/05/2023]
|
182
|
Konate K, Crombez L, Deshayes S, Decaffmeyer M, Thomas A, Brasseur R, Aldrian G, Heitz F, Divita G. Insight into the cellular uptake mechanism of a secondary amphipathic cell-penetrating peptide for siRNA delivery. Biochemistry 2010; 49:3393-402. [PMID: 20302329 DOI: 10.1021/bi901791x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Delivery of siRNA remains a major limitation to their clinical application, and several technologies have been proposed to improve their cellular uptake. We recently described a peptide-based nanoparticle system for efficient delivery of siRNA into primary cell lines: CADY. CADY is a secondary amphipathic peptide that forms stable complexes with siRNA and improves their cellular uptake independently of the endosomal pathway. In the present work, we have combined molecular modeling, spectroscopy, and membrane interaction approaches in order to gain further insight into CADY/siRNA particle mechanism of interaction with biological membrane. We demonstrate that CADY forms stable complexes with siRNA and binds phospholipids tightly, mainly through electrostatic interactions. Binding to siRNA or phospholipids triggers a conformational transition of CADY from an unfolded state to an alpha-helical structure, thereby stabilizing CADY/siRNA complexes and improving their interactions with cell membranes. Therefore, we propose that CADY cellular membrane interaction is driven by its structural polymorphism which enables stabilization of both electrostatic and hydrophobic contacts with surface membrane proteoglycan and phospholipids.
Collapse
Affiliation(s)
- Karidia Konate
- Centre de Recherches de Biochimie Macromoléculaire, CRBM-CNRS, UMR-5237, UM1-UM2, Department of Molecular Biophysics and Therapeutics, University of Montpellier, 1919 Route de Mende, 34293 Montpellier, France
| | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Pardaxin permeabilizes vesicles more efficiently by pore formation than by disruption. Biophys J 2010; 98:576-85. [PMID: 20159154 DOI: 10.1016/j.bpj.2009.08.063] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Revised: 08/02/2009] [Accepted: 08/06/2009] [Indexed: 12/28/2022] Open
Abstract
Pardaxin is a 33-amino-acid neurotoxin from the Red Sea Moses sole Pardachirus marmoratus, whose mode of action shows remarkable sensitivity to lipid chain length and charge, although the effect of pH is unclear. Here we combine optical spectroscopy and dye release experiments with laser scanning confocal microscopy and natural abundance (13)C solid-state nuclear magnetic resonance to provide a more complete picture of how pardaxin interacts with lipids. The kinetics and efficiency of release of entrapped calcein is highly sensitive to pH. In vesicles containing zwitterionic lipids (PC), release occurs most rapidly at low pH, whereas in vesicles containing 20% anionic lipid (PG), release occurs most rapidly at high pH. Pardaxin forms stable or transient pores in PC vesicles that allow release of contents without loss of vesicle integrity, whereas the inclusion of PG promotes total vesicle collapse. In agreement with this, solid-state nuclear magnetic resonance reveals that pardaxin takes up a trans-membrane orientation in 14-O-PC/6-O-PC bicelles, whereas the inclusion of 14-0-PG restricts it to contacts with lipid headgroups, promoting membrane lysis. Pore formation in zwitterionic vesicles is more efficient than lysis of anionic vesicles, suggesting that electrostatic interactions may trap pardaxin in several suboptimal interconverting conformations on the membrane surface.
Collapse
|
184
|
Kirwan JP, Hodges RS. Critical interactions in the stability control region of tropomyosin. J Struct Biol 2010; 170:294-306. [PMID: 20144718 PMCID: PMC2856757 DOI: 10.1016/j.jsb.2010.01.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 01/28/2010] [Accepted: 01/28/2010] [Indexed: 11/16/2022]
Abstract
Our laboratory has recently described a stability control region in the two-stranded alpha-helical coiled-coil alpha-tropomyosin that accounts for overall protein stability but is not required for folding (Hodges et al., 2009). We have used a synthetic peptide approach to investigate three stability control sites within the stability control region (residues 97-118). Two of the sites, electrostatic cluster 1 (97-104, EELDRAQE) and electrostatic cluster 2 (112-118, KLEEAEK), feature sequences with unusually high charge density and the potential to form multiple intrachain and interchain salt bridges (ionic attractions). A third site (105-111, RLATALQ) features an e position Leu residue, an arrangement known previously to enhance coiled-coil stability modestly. A native peptide and seven peptide analogs of the tropomyosin sequence 85-119 were prepared by Fmoc solid-phase peptide synthesis. Thermal stability measurements by circular dichroism (CD) spectroscopy revealed the following T(m) values for the native peptide and three key analogs: 52.9 degrees C (Native), 46.0 degrees C (R101A), 45.3 degrees C (K112A/K118A), and 27.9 degrees C (L110A). The corresponding DeltaT(m) values for the analogs, relative to the native peptide, are -6.9 degrees C, -7.6 degrees C, and -25.0 degrees C, respectively. The dramatic contribution to stability made by L110e is three times greater than the contribution of either electrostatic cluster 1 or 2, likely resulting from a novel hydrophobic interaction not previously observed. These thermal stability results were corroborated by temperature profiling analyses using reversed-phase high-performance liquid chromatography (RP-HPLC). We believe that the combined contributions of the interactions within the three stability control sites are responsible for the effect of the stability control region in tropomyosin, with the Leu110e contribution being most critical.
Collapse
Affiliation(s)
- J. Paul Kirwan
- Program in Structural Biology and Biophysics, Department of Biochemistry and Molecular Genetics, University of Colorado Denver, School of Medicine, Aurora, CO 80045
| | - Robert S. Hodges
- Program in Structural Biology and Biophysics, Department of Biochemistry and Molecular Genetics, University of Colorado Denver, School of Medicine, Aurora, CO 80045
| |
Collapse
|
185
|
Ahmad B, Haq SK, Varshney A, Moosavi-Movahedi AA, Khan RH. Effect of trifluoroethanol on native and acid-induced states of glucose oxidase from Aspergillus niger. BIOCHEMISTRY (MOSCOW) 2010; 75:486-530. [DOI: 10.1134/s0006297910040139] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
186
|
Patel AB, Khumsupan P, Narayanaswami V. Pyrene fluorescence analysis offers new insights into the conformation of the lipoprotein-binding domain of human apolipoprotein E. Biochemistry 2010; 49:1766-75. [PMID: 20073510 DOI: 10.1021/bi901902e] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The C-terminal domain (CT) of apolipoprotein E (apoE), a critical protein involved in cholesterol transport in the plasma and brain, plays an important role in high-affinity lipoprotein binding. Although high-resolution structural information is available for the N-terminal domain of apoE, the structural organization of the CT (residues 201-299) is largely unknown. In this study, we employ site-specific fluorescence labeling with pyrene maleimide to gain insight into the structure and conformation of apoE CT in its naturally self-associated state in buffer at physiologically relevant concentrations (5-50 microg/mL). Pyrene is a highly sensitive fluorophore that reports on spatial proximity between desired sites by displaying unique spectral features. Pyrene was covalently attached to single cysteine-containing recombinant human apoE CT at position 223 or 255 to probe the first predicted helical segment and at position 277 to monitor the terminal predicted helical segment. Regardless of the location of the probe, all three pyrene-labeled apoE CT variants display an intense and dramatic fluorescence excimer band at 460 nm, a signature feature of pyrene, which indicates that two pyrene moieties are within 10 A of each other. In addition, an intense peak at 387 nm (indicative of a highly hydrophobic environment) was noted in all cases. Fluorescence emission quenching by potassium iodide indicates that the accessibility to the probes was restricted at these locations. The possibility that the hydrophobicity of the pyrene moiety was the driving force for helix-helix interaction was excluded because pyrene located at position 209, which is predicted to be located in a nonhelical segment, did not display the above intense unique features. Lastly, denaturation studies suggest that the terminal helix unfolds prior to the first predicted helix in apoE CT. Our studies indicate that there are extensive intermolecular helix-helix contacts throughout the entire CT in the lipid-free state with two apoE CT molecules oriented parallel to each other to form a dimer, which dimerizes further to yield a tetramer. Such an organization allows helix-helix interactions to be replaced by helix-lipid interactions upon encountering a lipoprotein surface, with the terminal helix likely initiating the binding interaction. This study presents the possibility of employing pyrene fluorophores as powerful new alternatives to obtain conformational information of proteins at physiologically relevant concentrations.
Collapse
Affiliation(s)
- Arti B Patel
- Department of Chemistry and Biochemistry, 1250 Bellflower Boulevard, California State University Long Beach, Long Beach, California 90840, USA
| | | | | |
Collapse
|
187
|
Nagy A, Piszczek G, Sellers JR. Extensibility of the extended tail domain of processive and nonprocessive myosin V molecules. Biophys J 2010; 97:3123-31. [PMID: 20006949 DOI: 10.1016/j.bpj.2009.09.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 08/31/2009] [Accepted: 09/17/2009] [Indexed: 10/20/2022] Open
Abstract
Myosin V is a single-molecule motor that moves organelles along actin. When myosin V pulls loads inside the cell in a highly viscous environment, the force on the motor is unlikely to be constant. We propose that the tether between the single-molecule motor and the cargo (i.e., the extended tail domain of the molecule) must be able to absorb the sudden mechanical motions of the motor and allow smooth relaxation of the motion of the cargo to a new position. To test this hypothesis, we compared the elastic properties of the extended tail domains of processive (mouse myosin Va) and nonprocessive (Drosophila myosin V) molecular motors. The extended tail domain of these myosins consists of mechanically strong coiled-coil regions interspersed with flexible loops. In this work we explored the mechanical properties of coiled-coil regions using atomic force microscopy. We found that the processive and nonprocessive coiled-coil fragments display different unfolding patterns. The unfolding of coiled-coil structures occurs much later during the atomic force microscopy stretch cycle for processive myosin Va than for nonprocessive Drosophila myosin V, suggesting that this elastic tether between the cargo and motor may play an important role in sustaining the processive motions of this single-molecule motor.
Collapse
Affiliation(s)
- Attila Nagy
- Laboratory of Molecular Physiology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | |
Collapse
|
188
|
Abstract
The rational design of artificial enzymes, either by applying physico-chemical intuition of protein structure and function or with the aid of computational methods, is a promising area of research with the potential to tremendously impact medicine, industrial chemistry and energy production. Designed proteins also provide a powerful platform for dissecting enzyme mechanisms of natural systems. Artificial enzymes have come a long way from simple α-helical peptide catalysts to proteins that facilitate multistep chemical reactions designed by state-of-the-art computational methods. Looking forward, we examine strategies employed by natural enzymes that could be used to improve the speed and selectivity of artificial catalysts.
Collapse
Affiliation(s)
- Vikas Nanda
- Robert Wood Johnson Medical School - UMDNJ Biochemistry, Center for Advanced Biotechnology and Medicine, 679 Hoes Lane West, Piscataway, New Jersey 08854, USA.
| | | |
Collapse
|
189
|
Apostolovic B, Danial M, Klok HA. Coiled coils: attractive protein folding motifs for the fabrication of self-assembled, responsive and bioactive materials. Chem Soc Rev 2010; 39:3541-75. [DOI: 10.1039/b914339b] [Citation(s) in RCA: 223] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
190
|
Shiga D, Nakane D, Inomata T, Masuda H, Oda M, Noda M, Uchiyama S, Fukui K, Takano Y, Nakamura H, Mizuno T, Tanaka T. The effect of the side chain length of Asp and Glu on coordination structure of Cu(2+) in a de novo designed protein. Biopolymers 2009; 91:907-16. [PMID: 19598226 DOI: 10.1002/bip.21277] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Metal ions in proteins are important not only for the formation of the proper structures but also for various biological activities. For biological functions such as hydrolysis and oxidation, metal ions often adopt unusual coordination structures. We constructed a stable scaffold for metal binding to create distorted metal coordination structures. A stable four stranded alpha-helical coiled-coil structure was used as the scaffold, and the metal binding site was in the cavity created at the center of the structure. Two His residues and one Asp or Glu residue were used to coordinate the metal ions, AM2D and AM2E, respectively. Cu(2+) bound to AM2D with an equatorial planar coordination structure with two His, one Asp, and H(2)O as detected by electron spin resonance and UV spectral analyzes. On the other hand, Cu(2+) had a slightly distorted square planar structure when it bound two His and Glu in AM2E, due to the longer side-chain of the Glu residue as compared to the Asp residue. Computational analysis also supported the distorted coordination structure of Cu(2+) in AM2E. This construct should be useful to create various coordinations of metal ions for catalytic functions.
Collapse
Affiliation(s)
- Daigo Shiga
- Department of Material Sciences, Graduate School of Engineering, Nagoya Institute of Technology, Gokiso-chou, Nagoya 466-8555, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Hodges RS, Mills J, McReynolds S, Kirwan JP, Tripet B, Osguthorpe D. Identification of a unique "stability control region" that controls protein stability of tropomyosin: A two-stranded alpha-helical coiled-coil. J Mol Biol 2009; 392:747-62. [PMID: 19627992 PMCID: PMC2756485 DOI: 10.1016/j.jmb.2009.07.039] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 07/10/2009] [Accepted: 07/14/2009] [Indexed: 10/20/2022]
Abstract
Nine recombinant chicken skeletal alpha-tropomyosin proteins were prepared, eight C-terminal deletion constructs and the full length protein (1-81, 1-92, 1-99, 1-105, 1-110, 1-119, 1-131, 1-260 and 1-284) and characterized by circular dichroism spectroscopy and analytical ultracentrifugation. We identified for the first time, a stability control region between residues 97 and 118. Fragments of tropomyosin lacking this region (1-81, 1-92, and 1-99) still fold into two-stranded alpha-helical coiled-coils but are significantly less stable (T(m) between 26-28.5 degrees C) than longer fragments containing this region (1-119, 1-131, 1-260 and 1-284) which show a large increase in their thermal midpoints (T(m) 40-43 degrees C) for a DeltaT(m) of 16-18 degrees C between 1-99 and 1-119. We further investigated two additional fragments that ended between residues 99 and 119, that is fragments 1-105 and 1-110. These fragments were more stable than 1-99 and less stable than 1-119, and showed that there were three separate sites that synergistically contribute to the large jump in protein stability (electrostatic clusters 97-104 and 112-118, and a hydrophobic interaction from Leu 110). All the residues involved in these stabilizing interactions are located outside the hydrophobic core a and d positions that have been shown to be the major contributor to coiled-coil stability. Our results show clearly that protein stability is more complex than previously thought and unique sites can synergistically control protein stability over long distances.
Collapse
Affiliation(s)
- Robert S Hodges
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, 80045, USA.
| | | | | | | | | | | |
Collapse
|
192
|
Gondeau C, Corradin G, Heitz F, Le Peuch C, Balbo A, Schuck P, Kajava AV. The C-terminal domain of Plasmodium falciparum merozoite surface protein 3 self-assembles into alpha-helical coiled coil tetramer. Mol Biochem Parasitol 2009; 165:153-61. [PMID: 19428662 PMCID: PMC2680791 DOI: 10.1016/j.molbiopara.2009.01.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 01/28/2009] [Accepted: 01/29/2009] [Indexed: 11/19/2022]
Abstract
Proteins located on the surface of the pathogenic malaria parasite Plasmodium falciparum are objects of intensive studies due to their important role in the invasion of human cells and the accessibility to host antibodies thus making these proteins attractive vaccine candidates. One of these proteins, merozoite surface protein 3 (MSP3) represents a leading component among vaccine candidates; however, little is known about its structure and function. Our biophysical studies suggest that the 40 residue C-terminal domain of MSP3 protein self-assembles into a four-stranded alpha-helical coiled coil structure where alpha-helices are packed "side-by-side". A bioinformatics analysis provides an extended list of known and putative proteins from different species of Plasmodium which have such MSP3-like C-terminal domains. This finding allowed us to extend some conclusions of our studies to a larger group of the malaria surface proteins. Possible structural and functional roles of these highly conserved oligomerization domains in the intact merozoite surface proteins are discussed.
Collapse
Affiliation(s)
- Claire Gondeau
- Centre de Recherches de Biochimie Macromoléculaire, CNRS UMR-5237, University of Montpellier 1 and 2, Montpellier, France
| | | | - Frédéric Heitz
- Centre de Recherches de Biochimie Macromoléculaire, CNRS UMR-5237, University of Montpellier 1 and 2, Montpellier, France
| | - Christian Le Peuch
- Centre de Recherches de Biochimie Macromoléculaire, CNRS UMR-5237, University of Montpellier 1 and 2, Montpellier, France
| | - Andrea Balbo
- Dynamics of Macromolecular Assembly Section, Laboratory of Bioengineering and Physical Science, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, U.S.A
| | - Peter Schuck
- Dynamics of Macromolecular Assembly Section, Laboratory of Bioengineering and Physical Science, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, U.S.A
| | - Andrey V. Kajava
- Centre de Recherches de Biochimie Macromoléculaire, CNRS UMR-5237, University of Montpellier 1 and 2, Montpellier, France
| |
Collapse
|
193
|
Kish-Trier E, Wilkens S. Domain architecture of the stator complex of the A1A0-ATP synthase from Thermoplasma acidophilum. J Biol Chem 2009; 284:12031-40. [PMID: 19234304 PMCID: PMC2673272 DOI: 10.1074/jbc.m808962200] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Revised: 02/18/2009] [Indexed: 12/22/2022] Open
Abstract
A key structural element in the ion translocating F-, A-, and V-ATPases is the peripheral stalk, an assembly of two polypeptides that provides a structural link between the ATPase and ion channel domains. Previously, we have characterized the peripheral stalk forming subunits E and H of the A-ATPase from Thermoplasma acidophilum and demonstrated that the two polypeptides interact to form a stable heterodimer with 1:1 stoichiometry (Kish-Trier, E., Briere, L. K., Dunn, S. D., and Wilkens, S. (2008) J. Mol. Biol. 375, 673-685). To define the domain architecture of the A-ATPase peripheral stalk, we have now generated truncated versions of the E and H subunits and analyzed their ability to bind each other. The data show that the N termini of the subunits form an alpha-helical coiled-coil, approximately 80 residues in length, whereas the C-terminal residues interact to form a globular domain containingalpha- and beta-structure. We find that the isolated C-terminal domain of the E subunit exists as a dimer in solution, consistent with a recent crystal structure of the related Pyrococcus horikoshii A-ATPase E subunit (Lokanath, N. K., Matsuura, Y., Kuroishi, C., Takahashi, N., and Kunishima, N. (2007) J. Mol. Biol. 366, 933-944). However, upon the addition of a peptide comprising the C-terminal 21 residues of the H subunit (or full-length H subunit), dimeric E subunit C-terminal domain dissociates to form a 1:1 heterodimer. NMR spectroscopy was used to show that H subunit C-terminal peptide binds to E subunit C-terminal domain via the terminal alpha-helices, with little involvement of the beta-sheet region. Based on these data, we propose a structural model of the A-ATPase peripheral stalk.
Collapse
Affiliation(s)
- Erik Kish-Trier
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, New York 13210, USA
| | | |
Collapse
|
194
|
Legardinier S, Raguénès-Nicol C, Tascon C, Rocher C, Hardy S, Hubert JF, Le Rumeur E. Mapping of the lipid-binding and stability properties of the central rod domain of human dystrophin. J Mol Biol 2009; 389:546-58. [PMID: 19379759 DOI: 10.1016/j.jmb.2009.04.025] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 03/31/2009] [Accepted: 04/11/2009] [Indexed: 11/28/2022]
Abstract
Dystrophin is a cytoskeletal protein that confers resistance to the sarcolemma against the stress of contraction-relaxation cycles by interacting with cytoskeletal and membrane partners. Apart from several proteins, membrane phospholipids are a partner of the central rod domain made up of 24 spectrin-like repeats, separated into sub-domains by four hinges. We previously showed that repeats 1 to 3 bind to membrane anionic phospholipids, while repeats 20 to 24 are not able to do so. We focus here on the phospholipid-binding properties of the major part of the central rod domain, namely, the sub-domain delineated by hinges 2 and 3 comprising 16 repeats ranging from repeat 4 to 19 (R4-19). We designed and produced multirepeat proteins comprising three to five repeats and report their lipid-binding properties as well as their thermal stabilities. When these proteins are mixed with liposomes including the anionic lipid phosphatidylserine, they form stable protein-vesicle complexes as determined by gel-filtration chromatography. The absence of an anionic lipid precludes the formation of such complexes. Spectroscopic analyses by circular dichroism and tryptophan fluorescence show that, while the alpha-helical secondary structures are not modified by the binding, protein trans conformation leads to the movement of tryptophan residues into more hydrophobic environments. In addition, the decrease in the molar ellipticity ratio at 222/208 nm as observed by circular dichroism indicates that lipid binding reduces the inter-helical interactions of multirepeat proteins, thus suggesting partly "opened" coiled-coil structures. Combining these results with data from our previous studies, we propose a new model of the dystrophin molecule lying along the membrane bilayer, in which the two sub-domains R1-3 and R4-19 interact with lipids and F-actin, while the distal sub-domain R20-24 does not exhibit any interaction. These lipid-binding domains should thus maintain a structural link between cytoskeletal actin and sarcolemma via the membrane phospholipids.
Collapse
Affiliation(s)
- Sébastien Legardinier
- Université de Rennes 1, UMR CNRS 6026, Interactions cellulaires et moléculaires, IFR 140, Faculté de Médecine, CS 34317, 35043 Rennes Cedex, France
| | | | | | | | | | | | | |
Collapse
|
195
|
Two Distantly Spaced Basic Patches in the Flexible Domain of Huntingtin-Interacting Protein 1 (HIP1) Are Essential for the Binding of Clathrin Light Chain. Res Lett Biochem 2009; 2009:256124. [PMID: 22820750 PMCID: PMC3005887 DOI: 10.1155/2009/256124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2009] [Accepted: 02/24/2009] [Indexed: 11/17/2022] Open
Abstract
The interaction between HIP family proteins (HIP1 and HIP12/1R) and clathrin is fundamental to endocytosis. We used circular dichroism (CD) to study the stability of an HIP1 subfragment (aa468-530) that is splayed open. CD thermal melts show HIP1 468-530 is only stable at low temperatures, but this HIP1 fragment contains a structural unit that does not melt out even at 83°C. We then created HIP1 mutants to probe our hypothesis that a short hydrophobic path in the opened region is the binding site for clathrin light chain. We found that the binding of hub/LCb was sensitive to mutating two distantly separated basic residues (K474 and K494). The basic patches marked by K474 and K494 are conserved in HIP12/1R. The lack of conservation in sla2p (S. cerevisiae), HIP1 from D. melanogaster, and HIP1 homolog ZK370.3 from C. elegans implies the binding of HIP1 and HIP1 homologs to clathrin light chain may be different in these organisms.
Collapse
|
196
|
Peacock AFA, Iranzo O, Pecoraro VL. Harnessing natures ability to control metal ion coordination geometry using de novo designed peptides. Dalton Trans 2009:2271-80. [PMID: 19290357 PMCID: PMC3046812 DOI: 10.1039/b818306f] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Advances in protein chemistry and molecular and structural biology have empowered modern chemists to build complex biological architectures using a "first principles" approach, which is known as de novo protein design. In this Perspective we demonstrate how simple three-stranded alpha-helical constructs can be prepared by the sole consideration of the primary amino acid sequence of a peptide. With these well defined systems, we then demonstrate that metal binding cavities can be carved out of the hydrophobic cores of these aggregates in order to bind metal ions such as cadmium with well defined coordination geometries. Examples will be given of homoleptic CdS(3) complexes, CdS(3)O sites and proteins which contain equilibrium mixtures of these two species. We will provide a description of a strategy that allows us to build heterochromic peptides (small proteins that complex two metals in nearly identical environments but which result in different physical properties and allow for metal site selectivity). We conclude with a new class of designed peptides, diastereopeptides, which can exploit changes in amino acid chirality to control metal ion coordination number and lead to an alternative path towards heterochromic systems. The constructs described herein represent the initial steps of preparing protein structures that may simultaneous contain structural and catalytic metal binding centers. These studies inform the community on a developing field, which promises new opportunities for the study of bioinorganic chemistry.
Collapse
Affiliation(s)
- Anna F. A. Peacock
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109-1055
| | - Olga Iranzo
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109-1055
| | | |
Collapse
|
197
|
Afrin R, Takahashi I, Shiga K, Ikai A. Tensile mechanics of alanine-based helical polypeptide: force spectroscopy versus computer simulations. Biophys J 2009; 96:1105-14. [PMID: 19186146 DOI: 10.1016/j.bpj.2008.10.046] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Accepted: 10/27/2008] [Indexed: 11/16/2022] Open
Abstract
In nature, an alpha-helix is commonly used to build thermodynamically stable and mechanically rigid protein conformations. In view of growing interest in the mechanical rigidity of proteins, we measured the tensile profile of an alanine-based alpha-helical polypeptide on an atomic-force microscope to investigate the basic mechanics of helix extension with minimal interference from side-chain interactions. The peptide was extended to its maximum contour length with much less force than in reported cases of poly-L-Glu or poly-L-Lys, indicating that chain stiffness strongly depended on the physicochemical properties of side chains, such as their bulkiness. The low tensile-force extension originated presumably in locally unfolded parts because of spontaneous structural fluctuations. In 50% trifluoroethanol, the well-known helix-promoting agent, the rigidity of the sample polypeptide was markedly increased. Computer simulations of the peptide-stretching process showed that a majority of constituent residues underwent a transition from an alpha-helical to an extended conformation by overcoming an energy barrier around psi approximately 0 degrees on the Ramachandran plot. The observed lability of an isolated helix signified the biological importance of the lateral bundling of helices to maintain a rigid protein structure.
Collapse
Affiliation(s)
- Rehana Afrin
- Biofrontier Center, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | | | | | | |
Collapse
|
198
|
How methyl cyanide induces aggregation in all-alpha proteins: A case study in four albumins. Int J Biol Macromol 2009; 44:163-9. [DOI: 10.1016/j.ijbiomac.2008.11.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 11/22/2008] [Accepted: 11/24/2008] [Indexed: 11/30/2022]
|
199
|
Marenchino M, Armbruster DW, Hennig M. Rapid and efficient purification of RNA-binding proteins: application to HIV-1 Rev. Protein Expr Purif 2009; 63:112-9. [PMID: 18852051 PMCID: PMC2624574 DOI: 10.1016/j.pep.2008.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Revised: 09/04/2008] [Accepted: 09/10/2008] [Indexed: 10/21/2022]
Abstract
Non-specifically bound nucleic acid contaminants are an unwanted feature of recombinant RNA-binding proteins purified from Escherichia coli (E. coli). Removal of these contaminants represents an important step for the proteins' application in several biological assays and structural studies. The method described in this paper is a one-step protocol which is effective at removing tightly bound nucleic acids from overexpressed tagged HIV-1 Rev in E. coli. We combined affinity chromatography under denaturing conditions with subsequent on-column refolding, to prevent self-association of Rev while removing the nucleic acid contaminants from the end product. We compare this purification method with an established, multi-step protocol involving precipitation with polyethyleneimine (PEI). As our tailored protocol requires only one-step to simultaneously purify tagged proteins and eliminate bound cellular RNA and DNA, it represents a substantial advantage in time, effort, and expense.
Collapse
Affiliation(s)
- Marco Marenchino
- Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | - Mirko Hennig
- Medical University of South Carolina, Charleston, South Carolina 29425, USA
| |
Collapse
|
200
|
Sakurai Y, Mizuno T, Hiroaki H, Oku JI, Tanaka T. Optimization of aromatic side chain size complementarity in the hydrophobic core of a designed coiled-coil. ACTA ACUST UNITED AC 2008; 66:387-94. [PMID: 16316455 DOI: 10.1111/j.1399-3011.2005.00304.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The coiled-coil structure plays an important roles, especially in protein assembly. Previously we constructed AAB-type heterotrimeric coiled-coils by manipulating the packing in the hydrophobic core using Trp and Ala residues, where one Trp and two Ala residues were placed in the hydrophobic core instead of three Ile residues. To optimize the packing complementarity in the hydrophobic core, we investigated the effects of introducing various aromatic amino acids on the formation of an AAB-type heterotrimeric coiled-coil, by circular dichroism, thermal stability, and nuclear magnetic resonance (NMR) studies. We found that the Phe residue was more suitable for heterotrimeric coiled-coil formation than the Trp residue, when combined with two Ala residues, whereas the Tyr and His residues did not induce the coiled-coil structure efficiently.
Collapse
Affiliation(s)
- Y Sakurai
- Department of Material Sciences, Graduate School of Engineering, Nagoya Institute of Technology, Gokiso-chou, Nagoya 466-8555, Japan
| | | | | | | | | |
Collapse
|