1
|
McGowan J, Peter C, Chattopadhyay S, Chakravarti R. 14-3-3ζ-A Novel Immunogen Promotes Inflammatory Cytokine Production. Front Immunol 2019; 10:1553. [PMID: 31396202 PMCID: PMC6667649 DOI: 10.3389/fimmu.2019.01553] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/21/2019] [Indexed: 01/13/2023] Open
Abstract
The presence of autoantibodies against 14-3-3ζ in human autoimmune diseases indicates its antigenic function. However, neither the cause nor the consequence of this newly-identified antigenic function of 14-3-3ζ protein is known. To address this, we investigated the immunological functions of 14-3-3ζ by studying ex vivo effects on human peripheral blood mononuclear cells (PBMC) proliferation, polarization, and cytokine production. Exogenous 14-3-3ζ promoted PBMC proliferation and T cell polarization toward Th1 and Th17 populations. Significant increases in IFN-γ and IL-17 levels were observed in the presence of 14-3-3ζ. A specific increase in Th1 cells and IFN-γ production provided strong evidence for MHC class II presentation of 14-3-3ζ antigen. Particularly HLA-DRB1*0401 allele strongly promoted 14-3-3ζ-induced IFN-γ producing cells. In contrast, prednisolone treatment suppressed both 14-3-3ζ-induced T cell polarization and cytokine production. Overall, we show that MHC presentation and the adaptor functions of 14-3-3ζ participate in promoting IFN-γ and IL-17 production, two of the cytokines commonly associated with autoimmune diseases. To the best of our knowledge, this is the first report describing the ex vivo antigenic function of 14-3-3ζ with human PBMC, thereby providing the basis of its immunological role in human diseases.
Collapse
Affiliation(s)
- Jenna McGowan
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Cara Peter
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Saurabh Chattopadhyay
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Ritu Chakravarti
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| |
Collapse
|
2
|
Yan Y, Wang W, Sun Z, Zhang JZH, Ji C. Protein-Ligand Empirical Interaction Components for Virtual Screening. J Chem Inf Model 2017; 57:1793-1806. [PMID: 28678484 DOI: 10.1021/acs.jcim.7b00017] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A major shortcoming of empirical scoring functions is that they often fail to predict binding affinity properly. Removing false positives of docking results is one of the most challenging works in structure-based virtual screening. Postdocking filters, making use of all kinds of experimental structure and activity information, may help in solving the issue. We describe a new method based on detailed protein-ligand interaction decomposition and machine learning. Protein-ligand empirical interaction components (PLEIC) are used as descriptors for support vector machine learning to develop a classification model (PLEIC-SVM) to discriminate false positives from true positives. Experimentally derived activity information is used for model training. An extensive benchmark study on 36 diverse data sets from the DUD-E database has been performed to evaluate the performance of the new method. The results show that the new method performs much better than standard empirical scoring functions in structure-based virtual screening. The trained PLEIC-SVM model is able to capture important interaction patterns between ligand and protein residues for one specific target, which is helpful in discarding false positives in postdocking filtering.
Collapse
Affiliation(s)
- Yuna Yan
- Shanghai Engineering Research Center for Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University , Shanghai 200062, China.,State Key Laboratory of Precision Spectroscopy, East China Normal University , Shanghai 200062, China.,NYU-ECNU Center for Computational Chemistry at NYU Shanghai , Shanghai 200062, China
| | - Weijun Wang
- Shanghai Engineering Research Center for Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University , Shanghai 200062, China.,State Key Laboratory of Precision Spectroscopy, East China Normal University , Shanghai 200062, China.,NYU-ECNU Center for Computational Chemistry at NYU Shanghai , Shanghai 200062, China
| | - Zhaoxi Sun
- Shanghai Engineering Research Center for Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University , Shanghai 200062, China.,State Key Laboratory of Precision Spectroscopy, East China Normal University , Shanghai 200062, China.,NYU-ECNU Center for Computational Chemistry at NYU Shanghai , Shanghai 200062, China
| | - John Z H Zhang
- Shanghai Engineering Research Center for Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University , Shanghai 200062, China.,State Key Laboratory of Precision Spectroscopy, East China Normal University , Shanghai 200062, China.,NYU-ECNU Center for Computational Chemistry at NYU Shanghai , Shanghai 200062, China
| | - Changge Ji
- Shanghai Engineering Research Center for Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University , Shanghai 200062, China.,State Key Laboratory of Precision Spectroscopy, East China Normal University , Shanghai 200062, China.,NYU-ECNU Center for Computational Chemistry at NYU Shanghai , Shanghai 200062, China
| |
Collapse
|
3
|
Gurung AB, Das AK, Bhattacharjee A. Disruption of redox catalytic functions of peroxiredoxin-thioredoxin complex in Mycobacterium tuberculosis H37Rv using small interface binding molecules. Comput Biol Chem 2017; 67:69-83. [DOI: 10.1016/j.compbiolchem.2016.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 09/19/2016] [Accepted: 12/30/2016] [Indexed: 10/20/2022]
|
4
|
Grimme D, González-ruiz D, Gohlke* H. Computational Strategies and Challenges for Targeting Protein–Protein Interactions with Small Molecules. PHYSICO-CHEMICAL AND COMPUTATIONAL APPROACHES TO DRUG DISCOVERY 2012. [DOI: 10.1039/9781849735377-00319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
5
|
Fernández‐Recio J. Prediction of protein binding sites and hot spots. WILEY INTERDISCIPLINARY REVIEWS-COMPUTATIONAL MOLECULAR SCIENCE 2011. [DOI: 10.1002/wcms.45] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
6
|
McFadden MJ, Junop MS, Brennan JD. Magnetic “Fishing” Assay To Screen Small-Molecule Mixtures for Modulators of Protein−Protein Interactions. Anal Chem 2010; 82:9850-7. [PMID: 21067198 DOI: 10.1021/ac102164d] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Meghan J. McFadden
- Chemical Biology Graduate Program, Department of Chemistry and Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4M1, and Department of Biochemistry and Biomedical Engineering, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | - Murray S. Junop
- Chemical Biology Graduate Program, Department of Chemistry and Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4M1, and Department of Biochemistry and Biomedical Engineering, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | - John D. Brennan
- Chemical Biology Graduate Program, Department of Chemistry and Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4M1, and Department of Biochemistry and Biomedical Engineering, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| |
Collapse
|
7
|
Grosdidier S, Totrov M, Fernández-Recio J. Computer applications for prediction of protein-protein interactions and rational drug design. Adv Appl Bioinform Chem 2009; 2:101-23. [PMID: 21918619 PMCID: PMC3169948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
In recent years, protein-protein interactions are becoming the object of increasing attention in many different fields, such as structural biology, molecular biology, systems biology, and drug discovery. From a structural biology perspective, it would be desirable to integrate current efforts into the structural proteomics programs. Given that experimental determination of many protein-protein complex structures is highly challenging, and in the context of current high-performance computational capabilities, different computer tools are being developed to help in this task. Among them, computational docking aims to predict the structure of a protein-protein complex starting from the atomic coordinates of its individual components, and in recent years, a growing number of docking approaches are being reported with increased predictive capabilities. The improvement of speed and accuracy of these docking methods, together with the modeling of the interaction networks that regulate the most critical processes in a living organism, will be essential for computational proteomics. The ultimate goal is the rational design of drugs capable of specifically inhibiting or modifying protein-protein interactions of therapeutic significance. While rational design of protein-protein interaction inhibitors is at its very early stage, the first results are promising.
Collapse
Affiliation(s)
- Solène Grosdidier
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Spain
| | | | - Juan Fernández-Recio
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Spain,Correspondence: Juan Fernandez-Recio, Life Sciences Department, Barcelona Supercomputing Center, C/Jordi Girona 29, 08034 Barcelona, Spain, Tel +34 934137729, Fax +34 934137721, Email
| |
Collapse
|
8
|
Wawrzycka-Gorczyca I, Rzeszotarska B, Dżygiel A, Masiukiewicz E, Kozioł AE. Molecular and crystal structure of 5-amino-1H-[1,2,4]triazole-3-carboxylic acid derivatives. Z KRIST-CRYST MATER 2009. [DOI: 10.1524/zkri.218.7.480.20717] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Abstract
The analysis of molecular structures of three derivatives: 5-amino-3-carbomethoxy-1-methyl-1H-[1,2,4]-triazole (1), 1-acetyl-5-amino-3-carbomethoxy-1H-[1,2,4]-triazole (2) and 5-acetylamino-3-carbomethoxy-1-methyl-1H-[1,2,4]triazole (3) indicate the sensitivity of the N2–N1–C5 fragment of the triazole ring to the substitution. Molecular conformations are influenced by several intra- and intermolecular contacts. In all structures very extensive N–H···N/O and C–H(methyl)···O/N net of secondary interactions is formed. This kind of interactions is common in crystal structures which are proton-deficient and for this reason various types of multi-point interactions are forced. Besides, triazole π···π stacking interactions are observed.
Collapse
|
9
|
Abstract
The inhibition of protein-protein interactions using small molecules is a viable approach for the treatment of a range of pathological conditions that result from a malfunctioning of these interactions. Our strategy for the design of such agents involves the mimicry of side-chain residues on one face of the alpha-helix; these residues frequently play a key role in mediating protein-protein interactions. The first-generation terphenyl scaffold, with a 3,2',2''-substitution pattern, is able to successfully mimic key helix residues and disrupt therapeutically relevant interactions, including the Bcl-X(L)-Bak and the p53-hDM2 (human double minute 2) interactions that are implicated in cancer. The second- and third-generation scaffolds have resulted in greater synthetic accessibility and more drug-like character in these molecules.
Collapse
|
10
|
Babaoglu K, Simeonov A, Irwin JJ, Nelson ME, Feng B, Thomas CJ, Cancian L, Costi MP, Maltby DA, Jadhav A, Inglese J, Austin CP, Shoichet BK. Comprehensive mechanistic analysis of hits from high-throughput and docking screens against beta-lactamase. J Med Chem 2008; 51:2502-11. [PMID: 18333608 PMCID: PMC2655312 DOI: 10.1021/jm701500e] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
High-throughput screening (HTS) is widely used in drug discovery. Especially for screens of unbiased libraries, false positives can dominate "hit lists"; their origins are much debated. Here we determine the mechanism of every active hit from a screen of 70,563 unbiased molecules against beta-lactamase using quantitative HTS (qHTS). Of the 1,274 initial inhibitors, 95% were detergent-sensitive and were classified as aggregators. Among the 70 remaining were 25 potent, covalent-acting beta-lactams. Mass spectra, counter-screens, and crystallography identified 12 as promiscuous covalent inhibitors. The remaining 33 were either aggregators or irreproducible. No specific reversible inhibitors were found. We turned to molecular docking to prioritize molecules from the same library for testing at higher concentrations. Of 16 tested, 2 were modest inhibitors. Subsequent X-ray structures corresponded to the docking prediction. Analog synthesis improved affinity to 8 microM. These results suggest that it may be the physical behavior of organic molecules, not their reactivity, that accounts for most screening artifacts. Structure-based methods may prioritize weak-but-novel chemotypes in unbiased library screens.
Collapse
Affiliation(s)
- Kerim Babaoglu
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158-2330
| | - Anton Simeonov
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892-3370
| | - John J. Irwin
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158-2330
| | - Michael E. Nelson
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892-3370
| | - Brian Feng
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158-2330
| | - Craig J. Thomas
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892-3370
| | - Laura Cancian
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Modena e Reggio Emilia, Via Campi 183, 41100, Modena, Italy
| | - M. Paola Costi
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Modena e Reggio Emilia, Via Campi 183, 41100, Modena, Italy
| | - David A. Maltby
- Mass Spectroscopy Facility, Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143-0446
| | - Ajit Jadhav
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892-3370
| | - James Inglese
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892-3370
| | - Christopher P. Austin
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892-3370
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158-2330
| |
Collapse
|
11
|
Graves AP, Shivakumar DM, Boyce SE, Jacobson MP, Case DA, Shoichet BK. Rescoring docking hit lists for model cavity sites: predictions and experimental testing. J Mol Biol 2008; 377:914-34. [PMID: 18280498 PMCID: PMC2752715 DOI: 10.1016/j.jmb.2008.01.049] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2007] [Revised: 01/12/2008] [Accepted: 01/17/2008] [Indexed: 01/07/2023]
Abstract
Molecular docking computationally screens thousands to millions of organic molecules against protein structures, looking for those with complementary fits. Many approximations are made, often resulting in low "hit rates." A strategy to overcome these approximations is to rescore top-ranked docked molecules using a better but slower method. One such is afforded by molecular mechanics-generalized Born surface area (MM-GBSA) techniques. These more physically realistic methods have improved models for solvation and electrostatic interactions and conformational change compared to most docking programs. To investigate MM-GBSA rescoring, we re-ranked docking hit lists in three small buried sites: a hydrophobic cavity that binds apolar ligands, a slightly polar cavity that binds aryl and hydrogen-bonding ligands, and an anionic cavity that binds cationic ligands. These sites are simple; consequently, incorrect predictions can be attributed to particular errors in the method, and many likely ligands may actually be tested. In retrospective calculations, MM-GBSA techniques with binding-site minimization better distinguished the known ligands for each cavity from the known decoys compared to the docking calculation alone. This encouraged us to test rescoring prospectively on molecules that ranked poorly by docking but that ranked well when rescored by MM-GBSA. A total of 33 molecules highly ranked by MM-GBSA for the three cavities were tested experimentally. Of these, 23 were observed to bind--these are docking false negatives rescued by rescoring. The 10 remaining molecules are true negatives by docking and false positives by MM-GBSA. X-ray crystal structures were determined for 21 of these 23 molecules. In many cases, the geometry prediction by MM-GBSA improved the initial docking pose and more closely resembled the crystallographic result; yet in several cases, the rescored geometry failed to capture large conformational changes in the protein. Intriguingly, rescoring not only rescued docking false positives, but also introduced several new false positives into the top-ranking molecules. We consider the origins of the successes and failures in MM-GBSA rescoring in these model cavity sites and the prospects for rescoring in biologically relevant targets.
Collapse
Affiliation(s)
- Alan P. Graves
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 1700 4th Street, San Francisco, CA 94158-2330, USA
- Graduate Group in Biophysics, University of California, San Francisco, 1700 4th Street, San Francisco, CA 94158-2330, USA
| | - Devleena M. Shivakumar
- Department of Molecular Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sarah E. Boyce
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 1700 4th Street, San Francisco, CA 94158-2330, USA
- Graduate Group in Chemistry and Chemical Biology, University of California, San Francisco, 1700 4th Street, San Francisco, CA 94158-2330, USA
| | - Matthew P. Jacobson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 1700 4th Street, San Francisco, CA 94158-2330, USA
| | - David A. Case
- Department of Molecular Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 1700 4th Street, San Francisco, CA 94158-2330, USA
| |
Collapse
|
12
|
Feng JN, Yu ZY, He XH, Shen BF. A5, a new small-molecule inhibitor of CD4 D1 obtained from a computer-aided screening method, contributes to the inhibition of CD4+ T-cell function. ACTA ACUST UNITED AC 2007; 12:800-8. [PMID: 17644770 DOI: 10.1177/1087057107305505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In this study, the authors apply a computer-based strategy to screen thousands of small-molecule, nonpeptidic organic compounds in the Available Chemicals Directory database and to select a series of potential candidates as ligands of the proposed CD4 D1 surface pocket. Then, several cell-based models are used to determine the actual biological functions of these compounds. A small molecule designated A5 (N-((pyridine-4-yl)methylene)thiophene-2-carbohydrazide) was obtained by a virtual screening followed by 3 cell-based functional assays. The results show that A5 could specifically block the CD4-major histocompatibility complex II binding in a rosetting assay, inhibit the mixed lymphocyte reaction-induced T-cell proliferation in a concentration-dependent manner, and reduce the PMA plus ionomycin-stimulated interleukin-2 secretion from peripheral blood mononuclear cells.
Collapse
|
13
|
Abstract
Ligand enrichment among top-ranking hits is a key metric of molecular docking. To avoid bias, decoys should resemble ligands physically, so that enrichment is not simply a separation of gross features, yet be chemically distinct from them, so that they are unlikely to be binders. We have assembled a directory of useful decoys (DUD), with 2950 ligands for 40 different targets. Every ligand has 36 decoy molecules that are physically similar but topologically distinct, leading to a database of 98,266 compounds. For most targets, enrichment was at least half a log better with uncorrected databases such as the MDDR than with DUD, evidence of bias in the former. These calculations also allowed 40x40 cross-docking, where the enrichments of each ligand set could be compared for all 40 targets, enabling a specificity metric for the docking screens. DUD is freely available online as a benchmarking set for docking at http://blaster.docking.org/dud/.
Collapse
Affiliation(s)
- Niu Huang
- Department of Pharmaceutical Chemistry, University of California San Francisco, QB3 Building, 1700 4th Street, Box 2550, San Francisco, California 94143-2550, USA
| | | | | |
Collapse
|
14
|
Yin H, Hamilton AD. Strategies for targeting protein-protein interactions with synthetic agents. Angew Chem Int Ed Engl 2006; 44:4130-63. [PMID: 15954154 DOI: 10.1002/anie.200461786] [Citation(s) in RCA: 382] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The development of small-molecule modulators of protein-protein interactions is a formidable goal, albeit one that possesses significant potential for the discovery of novel therapeutics. Despite the daunting challenges, a variety of examples exists for the inhibition of two large protein partners with low-molecular-weight ligands. This review discusses the strategies for targeting protein-protein interactions and the state of the art in the rational design of molecules that mimic the structures and functions of their natural targets.
Collapse
Affiliation(s)
- Hang Yin
- Yale University, New Haven, CT, USA
| | | |
Collapse
|
15
|
Massa SM, Xie Y, Yang T, Harrington AW, Kim ML, Yoon SO, Kraemer R, Moore LA, Hempstead BL, Longo FM. Small, nonpeptide p75NTR ligands induce survival signaling and inhibit proNGF-induced death. J Neurosci 2006; 26:5288-300. [PMID: 16707781 PMCID: PMC6675309 DOI: 10.1523/jneurosci.3547-05.2006] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Studies showing that neurotrophin binding to p75NTR can promote cell survival in the absence of Trk (tropomyosin-related kinase) receptors, together with recent structural data indicating that NGF may bind to p75NTR in a monovalent manner, raise the possibility that small molecule p75NTR ligands that positively regulate survival might be found. A pharmacophore designed to capture selected structural and physical chemical features of a neurotrophin domain known to interact with p75NTR was applied to in silico screening of small molecule libraries. Small, nonpeptide, monomeric compounds were identified that interact with p75NTR. In cells showing trophic responses to neurotrophins, the compounds promoted survival signaling through p75NTR-dependent mechanisms. In cells susceptible to proneurotrophin-induced death, compounds did not induce apoptosis but inhibited proneurotrophin-mediated death. These studies identify a unique range of p75NTR behaviors that can result from isolated receptor liganding and establish several novel therapeutic leads.
Collapse
Affiliation(s)
- Stephen M. Massa
- Correspondence should be addressed to either of the following: Dr. Frank M. Longo, Department of Neurology and Neurological Sciences A343, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, or Dr. Stephen M. Massa, Department of Neurology (127), San Francisco Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA 94121,
| | - Youmei Xie
- Correspondence should be addressed to either of the following: Dr. Frank M. Longo, Department of Neurology and Neurological Sciences A343, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, or Dr. Stephen M. Massa, Department of Neurology (127), San Francisco Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA 94121,
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Moldovan MC, Sabbagh L, Breton G, Sékaly RP, Krummel MF. Triggering of T cell activation via CD4 dimers. THE JOURNAL OF IMMUNOLOGY 2006; 176:5438-45. [PMID: 16622011 DOI: 10.4049/jimmunol.176.9.5438] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The onset of activation in Th cells is triggered by localized co-engagement of TCRs and the coreceptor CD4. A CD4 crystal suggested that CD4 may form dimers in some circumstances. In this study, we use live-cell fluorescence resonance energy transfer imaging to demonstrate that CD4 dimers are present at a basal level on the cell surface and accumulate at the synapse. Mechanistically, we reveal two conditions under which dimers are highly relevant. First, CD4 dimers are more proficient in mediating prolonged cell contacts with APCs in the presence or absence of Ag. This is consistent with a model whereby the dimer functions to increase T-APC avidity. Second, we show that dimer mutations result in an increased level of an inactive lckTyr(505) bound to the CD4 molecule relative to dimer-competent CD4. We also find a consistent defect in signaling onset in these cells. This supports a role for CD4 dimerization in maintaining active signaling machinery. We suggest that modulation of the dimer/monomer ratio may permit tuning of activation thresholds during initial engagement.
Collapse
|
17
|
Yin H, Hamilton AD. Strategien zur Modulation von Protein-Protein-Wechselwirkungen mit synthetischen Substanzen. Angew Chem Int Ed Engl 2005. [DOI: 10.1002/ange.200461786] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
18
|
Roy CJ, Warfield KL, Welcher BC, Gonzales RF, Larsen T, Hanson J, David CS, Krakauer T, Bavari S. Human leukocyte antigen-DQ8 transgenic mice: a model to examine the toxicity of aerosolized staphylococcal enterotoxin B. Infect Immun 2005; 73:2452-60. [PMID: 15784591 PMCID: PMC1087414 DOI: 10.1128/iai.73.4.2452-2460.2005] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcal enterotoxins (SEs) belong to a large group of bacterial exotoxins that cause severe immunopathologies, especially when delivered as an aerosol. SEs elicit the release of lethal amounts of cytokines by binding to major histocompatibility complex (MHC) class II and cross-linking susceptible T-cell receptors. Efforts to develop effective therapeutic strategies to protect against SEs delivered as an aerosol have been hampered by the lack of small animal models that consistently emulate human responses to these toxins. Here, we report that human leukocyte antigen-DQ8 (HLA-DQ8) transgenic (Tg) mice, but not littermate controls, succumbed to lethal shock induced by SEB aerosols without potentiation. Substantial amounts of perivascular edema and inflammatory infiltrates were noted in the lungs of Tg mice, similar to the pathology observed in nonhuman primates exposed by aerosol to SEB. Furthermore, the observed pathologies and lethal shock correlated with an upsurge in proinflammatory cytokine mRNA gene expression in the lungs and spleens, as well as with marked increases in the levels of proinflammatory circulating cytokines in the Tg mice. Unlike the case for littermate controls, telemetric evaluation showed significant hypothermia in Tg mice exposed to lethal doses of SEB. Taken together, these results show that this murine model will allow for the examination of therapeutics and vaccines developed specifically against SEB aerosol exposure and possibly other bacterial superantigens in the context of human MHC class II receptors.
Collapse
Affiliation(s)
- Chad J Roy
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Gohlke H, Kiel C, Case DA. Insights into protein-protein binding by binding free energy calculation and free energy decomposition for the Ras-Raf and Ras-RalGDS complexes. J Mol Biol 2003; 330:891-913. [PMID: 12850155 DOI: 10.1016/s0022-2836(03)00610-7] [Citation(s) in RCA: 1008] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Absolute binding free energy calculations and free energy decompositions are presented for the protein-protein complexes H-Ras/C-Raf1 and H-Ras/RalGDS. Ras is a central switch in the regulation of cell proliferation and differentiation. In our study, we investigate the capability of the molecular mechanics (MM)-generalized Born surface area (GBSA) approach to estimate absolute binding free energies for the protein-protein complexes. Averaging gas-phase energies, solvation free energies, and entropic contributions over snapshots extracted from trajectories of the unbound proteins and the complexes, calculated binding free energies (Ras-Raf: -15.0(+/-6.3)kcal mol(-1); Ras-RalGDS: -19.5(+/-5.9)kcal mol(-1)) are in fair agreement with experimentally determined values (-9.6 kcal mol(-1); -8.4 kcal mol(-1)), if appropriate ionic strength is taken into account. Structural determinants of the binding affinity of Ras-Raf and Ras-RalGDS are identified by means of free energy decomposition. For the first time, computationally inexpensive generalized Born (GB) calculations are applied in this context to partition solvation free energies along with gas-phase energies between residues of both binding partners. For selected residues, in addition, entropic contributions are estimated by classical statistical mechanics. Comparison of the decomposition results with experimentally determined binding free energy differences for alanine mutants of interface residues yielded correlations with r(2)=0.55 and 0.46 for Ras-Raf and Ras-RalGDS, respectively. Extension of the decomposition reveals residues as far apart as 25A from the binding epitope that can contribute significantly to binding free energy. These "hotspots" are found to show large atomic fluctuations in the unbound proteins, indicating that they reside in structurally less stable regions. Furthermore, hotspot residues experience a significantly larger-than-average decrease in local fluctuations upon complex formation. Finally, by calculating a pair-wise decomposition of interactions, interaction pathways originating in the binding epitope of Raf are found that protrude through the protein structure towards the loop L1. This explains the finding of a conformational change in this region upon complex formation with Ras, and it may trigger a larger structural change in Raf, which is considered to be necessary for activation of the effector by Ras.
Collapse
Affiliation(s)
- Holger Gohlke
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
20
|
Brive L, Abagyan R. Computational structural proteomics. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2002:149-66. [PMID: 12061000 DOI: 10.1007/978-3-662-04747-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- L Brive
- Department of Molecular Biology, TPC-28, Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | |
Collapse
|
21
|
Moldovan MC, Yachou A, Lévesque K, Wu H, Hendrickson WA, Cohen EA, Sékaly RP. CD4 dimers constitute the functional component required for T cell activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:6261-8. [PMID: 12444132 DOI: 10.4049/jimmunol.169.11.6261] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The CD4 molecule plays a key role in the development and activation of helper T cells. Dimerization and oligomerization is often a necessary step in the function of several cell surface receptors. Herein, we provide direct biochemical evidence confirming the presence of CD4 as dimers in transfected cells from hemopoetic and fibroblastic origin as well as in primary T cells. Such dimers are also observed with murine CD4 confirming selective pressure during evolution to maintain such a structure. Using a series of point mutations, we have precisely mapped the dimerization site at residues K318 and Q344 within the fourth extracellular domain of CD4. These residues are highly conserved and their mutation results in interference with dimer formation. More importantly, we demonstrate that dimer formation is essential for the coligand and coreceptor functions of CD4 in T cell activation. These data strongly suggest that CD4 dimerization is necessary for helper T cell function.
Collapse
|
22
|
Gao GF, Rao Z, Bell JI. Molecular coordination of alphabeta T-cell receptors and coreceptors CD8 and CD4 in their recognition of peptide-MHC ligands. Trends Immunol 2002; 23:408-13. [PMID: 12133804 DOI: 10.1016/s1471-4906(02)02282-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The interaction of the alphabeta T-cell receptor (TCR) with its ligand, peptide-MHC (pMHC), is enhanced by the recognition of the coreceptor CD8 or CD4 to the same pMHC in the immunological synapse. In the past few years, the coordination of these interactions at the molecular level has been revealed by analysis of their complex crystal structures and binding dynamics. Here we discuss the interactions of pMHC with the TCR and coreceptor CD8 or CD4 on the surfaces of alphabeta T cells and antigen presenting cells, and the implications for TCR signalling and the T-cell repertoire.
Collapse
Affiliation(s)
- George F Gao
- Nuffield Dept. of Clinical Medicine, John Radcliffe Hospital, University of Oxford, Headington, Oxford, UK.
| | | | | |
Collapse
|
23
|
Chapman RL, Stanley TB, Hazen R, Garvey EP. Small molecule modulators of HIV Rev/Rev response element interaction identified by random screening. Antiviral Res 2002; 54:149-62. [PMID: 12062388 DOI: 10.1016/s0166-3542(01)00222-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A high throughput scintillation proximity assay with biotinylated human immunodeficiency virus (HIV) Rev protein and tritiated Rev response element RNA was used to screen over 500,000 small molecules. Several chemical classes of inhibitors and two chemical classes of enhancers of binding were identified, with the molecular weight range being 400-600. The most common structural motif of inhibitor was an acidic moiety at the end of a linear aromatic system. Most of these modulators had EC(50) values in the 1-10 microM potency range, with several below 1 microM. Several classes displayed structure-activity relationships suggesting specific molecular interactions between small molecule and macromolecule. Several molecules were confirmed as inhibitors in a gel shift assay and by surface plasmon resonance analysis. Furthermore, one inhibitor was shown to bind the Rev protein with a binding constant equal to its IC(50) value, consistent with the mechanism of inhibition being binding Rev. Thus, small molecules can modulate this macromolecular protein-RNA interaction in vitro. However, no compound demonstrated HIV antiviral activity in a relevant cell-based assay.
Collapse
Affiliation(s)
- Richard L Chapman
- Department of Molecular Screening, GlaxoSmithKline, PO Box 1-3398, Research Triangle Park, NC 27709-3398, USA
| | | | | | | |
Collapse
|
24
|
Park HS, Lin Q, Hamilton AD. Modulation of protein-protein interactions by synthetic receptors: design of molecules that disrupt serine protease-proteinaceous inhibitor interaction. Proc Natl Acad Sci U S A 2002; 99:5105-9. [PMID: 11959960 PMCID: PMC122729 DOI: 10.1073/pnas.082675899] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
In the present article we describe the design and evaluation of a synthetic receptor that binds to the exterior surface of chymotrypsin and disrupts its interaction with proteinaceous inhibitors, such as soybean trypsin inhibitor, basic pancreatic trypsin inhibitor, ovomucoid turkey inhibitor, and Bowman-Birk inhibitor. Using enzyme kinetics, nondenaturing gel electrophoresis, and gel filtration chromatography we show that the receptor is particularly effective at blocking the chymotrypsin-soybean trypsin inhibitor complex and that the mechanism involves formation of an initial ternary complex followed by a time-dependent displacement of the proteinaceous inhibitor.
Collapse
Affiliation(s)
- Hyung Soon Park
- Department of Chemistry, Yale University, New Haven, CT, 06520, USA
| | | | | |
Collapse
|
25
|
Edling AE, Choksi S, Huang Z, Korngold R. An organic CD4 inhibitor reduces the clinical and pathological symptoms of acute experimental allergic encephalomyelitis. J Autoimmun 2002; 18:169-79. [PMID: 11908949 DOI: 10.1006/jaut.2001.0576] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
CD4(+) T cells have an important role in mediating the pathogenesis of many human and experimental autoimmune diseases including experimental allergic encephalomyelitis (EAE), a demyelinating animal model for multiple sclerosis (MS). We applied a computer screening approach to select a small organic molecule, TJU103, that would specifically inhibit autoreactive CD4(+) T cells by disrupting the function of the CD4 molecule during activation. Upon studying the therapeutic effect of TJU103 in acute EAE, it was found that administration shortly before or after the onset of clinical symptoms reduced the severity of disease in both SJL and SWXJ-14 mouse models. In addition, TJU103 treatment could affect both in vivo responses to EAE rechallenge and secondary in vitro proliferation and cytokine production of T cells responding to proteolipid protein epitope 139-151 (PLPe). These results demonstrate the potential of the TJU103 organic inhibitor for future clinical application in CD4(+) T cell-mediated diseases.
Collapse
Affiliation(s)
- Andrea E Edling
- Kimmel Cancer Institute, Jefferson Medical College, Philadelphia, PA 19107, USA
| | | | | | | |
Collapse
|
26
|
Ren Y, Feng J, Qu H, Li S, Shen B. Three-dimensional structure and function study on the active region in the extracellular ligand-binding domain of human IL-6 receptor. SCIENCE IN CHINA. SERIES C, LIFE SCIENCES 2000; 43:425-32. [PMID: 18726347 DOI: 10.1007/bf02879308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/1999] [Indexed: 11/26/2022]
Abstract
In this study the three-dimensional (3-D) model of the ligand-binding domain (V106-P322) of human interleukin-6 receptor (hIL-6 R) was constructed by computer-guided homology modeling technique using the crystal structure of the ligand-binding domain (K52-L251) of human growth hormone receptor (hGHR) as templet. Furthermore, the active binding region of the 3-D model of hIL-6R with the ligand (hIL-6) was predicted. In light of the structural characteristics of the active region, a hydrophobic pocket shielded by two hydrophilic residues (E115 and E505) of the region was identified by a combination of molecular modelling and the site-directed or double-site mutation of the twelve crucial residues in the ligand-binding domain of hIL-6R (V106-P322). We observed and analyzed the effects of these mutants on the spatial conformation of the pocket-like region of hIL-6 R. The results indicated that any site-directed mutation of the five Cys residues (four conservative Cys residues: Cys121, Cys132, Cys165, Cys176; near membrane Cys residue: Cys193) or each double-site mutation of the five residues in WSEWS motif of hIL-6R (V106-P322) makes the corresponding spatial conformation of the pocket region block the linkage between hIL-6 R and hIL-6. However, the influence of the site-directed mutation of Cys211 and Cys277 individually on the conformation of the pocket region benefits the interaction between hIL-6R and hIL-6. Our study suggests that the predicted hydrophobic pocket in the 3-D model of hIL-6R (V106-P322) is the critical molecular basis for the binding of hIL-6R with its ligand, and the active pocket may be used as a target for designing small hIL-6R-inhibiting molecules in our further study.
Collapse
Affiliation(s)
- Y Ren
- Institute of Basic Medical Sciences, Chinese Academy of Military Medical Sciences, Beijing, China.
| | | | | | | | | |
Collapse
|
27
|
Wang JL, Liu D, Zhang ZJ, Shan S, Han X, Srinivasula SM, Croce CM, Alnemri ES, Huang Z. Structure-based discovery of an organic compound that binds Bcl-2 protein and induces apoptosis of tumor cells. Proc Natl Acad Sci U S A 2000; 97:7124-9. [PMID: 10860979 PMCID: PMC16510 DOI: 10.1073/pnas.97.13.7124] [Citation(s) in RCA: 907] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bcl-2 and related proteins are key regulators of apoptosis or programmed cell death implicated in human disease including cancer. We recently showed that cell-permeable Bcl-2 binding peptides could induce apoptosis of human myeloid leukemia in vitro and suppress its growth in severe combined immunodeficient mice. Here we report the discovery of HA14-1, a small molecule (molecular weight = 409) and nonpeptidic ligand of a Bcl-2 surface pocket, by using a computer screening strategy based on the predicted structure of Bcl-2 protein. In vitro binding studies demonstrated the interaction of HA14-1 with this Bcl-2 surface pocket that is essential for Bcl-2 biological function. HA14-1 effectively induced apoptosis of human acute myeloid leukemia (HL-60) cells overexpressing Bcl-2 protein that was associated with the decrease in mitochondrial membrane potential and activation of caspase-9 followed by caspase-3. Cytokine response modifier A, a potent inhibitor of Fas-mediated apoptosis, did not block apoptosis induced by HA14-1. Whereas HA14-1 strongly induced the death of NIH 3T3 (Apaf-1(+/+)) cells, it had little apoptotic effect on Apaf-1-deficient (Apaf-1(-/-)) mouse embryonic fibroblast cells. These data are consistent with a mechanism by which HA14-1 induces the activation of Apaf-1 and caspases, possibly by binding to Bcl-2 protein and inhibiting its function. The discovery of this cell-permeable molecule provides a chemical probe to study Bcl-2-regulated apoptotic pathways in vivo and could lead to the development of new therapeutic agents.
Collapse
Affiliation(s)
- J L Wang
- Kimmel Cancer Center, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
We review the recent progress made in our laboratories in structure-based drug design targeting proteins of the immunoglobulin superfamily (IgSF). We will focus on the CD4 protein, which is involved in T cell function, as a specific example of how the general concept and methodologies can be applied. Recent studies of CD4 structure and function have revealed new insight into possible mechanisms for CD4 self-association and its role in binding to major histocompatibility complex (MHC) class II molecules and initiation of T cell activation. This has led to the formulation of a hypothetical model of co-oligomerization of CD4, MHC class II, and T cell receptor (TCR). Such a basic understanding of CD4 structure and mechanisms has aided the development of a new generation of potential immunotherapeutics targeting specific CD4 surface functional sites. The design and discovery of small molecular inhibitors of CD4 and other IgSF proteins, in peptide, peptidomimetic, and nonpeptidic organic forms have opened new avenues for chemical research in which peptide, organic, and more recently combinatorial chemistry techniques can be used to further develop these promising lead analogs into a new generation of effective pharmaceuticals.
Collapse
Affiliation(s)
- Z Huang
- Kimmel Cancer Institute, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | |
Collapse
|
29
|
Huang Z. Structural chemistry and therapeutic intervention of protein-protein interactions in immune response, human immunodeficiency virus entry, and apoptosis. Pharmacol Ther 2000; 86:201-15. [PMID: 10882809 DOI: 10.1016/s0163-7258(00)00052-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Protein-protein interactions involved in diverse biological functions are largely unexplored therapeutic targets, and present a major challenge and opportunity for drug design research. Encouraging new approaches to this problem recently have emerged from studies of small molecule regulators of protein-protein complexes. This review outlines the basic concepts for two of these approaches, based on structural and chemical strategies, by illustrating their application in the design of small molecule inhibitors for three biological systems: (1) cell surface molecules CD4 and CD8 involved in immune response, (2) chemokine receptor-ligand interactions implicated in human immunodeficiency virus entry, and (3) B-cell leukemia/lymphoma-2 family proteins essential for regulation of programmed cell death or apoptosis. The design and discovery of these novel reagents provide valuable tools to probe fundamental questions about a particular protein-protein complex, and may lead to a new generation of potential therapeutic agents. Furthermore, these studies suggest a framework for chemical intervention of other protein-protein interactions involved in many pathological processes.
Collapse
Affiliation(s)
- Z Huang
- Kimmel Cancer Institute, Jefferson Medical College, Thomas Jefferson University, 802 BLSB, 233 South 10th Street, Philadelphia, PA 19107 USA.
| |
Collapse
|
30
|
Luo P, Canziani G, Cunto-Amesty G, Kieber-Emmons T. A molecular basis for functional peptide mimicry of a carbohydrate antigen. J Biol Chem 2000; 275:16146-54. [PMID: 10748116 DOI: 10.1074/jbc.m909121199] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Peptides may substitute for carbohydrate antigens in carbohydrate-specific immunological reactions. Using the recognition properties of an anti-Lewis Y (LeY) antibody, BR55-2, as a model system, we establish a molecular perspective for peptide mimicry by comparing the three-dimensional basis of BR55-2 binding to LeY with the binding of the same antibody to peptides. The peptides compete with LeY, as demonstrated by enzyme-linked immunosorbent assay and Biacore analysis. The computer program LUDI was used to epitope map the antibody-combining site, correlating peptide reactivity patterns. This approach identified amino acids interacting with the same BR55-2 functional residue groups that recognize the Fucalpha(1-3) moiety of LeY. Molecular modeling indicates that the peptides adopt an extended turn conformation within the BR55-2 combining site, serving to overlap the peptides with the LeY spatial position. Peptide binding is associated with only minor changes in BR55-2, relative to the BR55-2-LeY complex. Anti-peptide serum distinguishes the Fucalpha(1-3) from the Fucalpha(1-4) linkage, therefore differentiating difucosylated neolactoseries antigens. These results further confirm that peptides and carbohydrates can bind to the same antibody-binding site and that peptides can structurally and functionally mimic salient features of carbohydrate epitopes.
Collapse
Affiliation(s)
- P Luo
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
31
|
Krakauer T. Coordinate suppression of superantigen-induced cytokine production and T-cell proliferation by a small nonpeptidic inhibitor of class II major histocompatibility complex and CD4 interaction. Antimicrob Agents Chemother 2000; 44:1067-9. [PMID: 10722514 PMCID: PMC89815 DOI: 10.1128/aac.44.4.1067-1069.2000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Proinflammatory cytokines mediate the toxic effect of superantigenic staphylococcal exotoxins (SE). TJU103, a small nonpeptidic molecule that blocks the interaction between major histocompatibility complex class II and CD4 molecules inhibited SE-stimulated T-cell proliferation (by 92%) and production of tumor necrosis factor, interleukin 1beta, interleukin 6, and gamma interferon (by 66, 56, 76, and 72%, respectively) by human peripheral blood mononuclear cells. These data suggest that TJU103 may be useful for mitigating the pathogenic effects of SE.
Collapse
Affiliation(s)
- T Krakauer
- Department of Immunology and Molecular Biology, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland 21702-5011, USA.
| |
Collapse
|
32
|
Blum JH, Dove SL, Hochschild A, Mekalanos JJ. Isolation of peptide aptamers that inhibit intracellular processes. Proc Natl Acad Sci U S A 2000; 97:2241-6. [PMID: 10688899 PMCID: PMC15785 DOI: 10.1073/pnas.040573397] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We have developed a method for isolation of random peptides that inhibit intracellular processes in bacteria. A library of random peptides expressed as fusions to Escherichia coli thioredoxin (aptamers) were expressed under the tight control of the arabinose-inducible P(BAD) promoter. A selection was applied to the library to isolate aptamers that interfered with the activity of thymidylate synthase (ThyA) in vivo. Expression of an aptamer isolated by this method resulted in a ThyA(-) phenotype that was suppressed by simultaneous overexpression of ThyA. Two-hybrid analysis showed that this aptamer is likely to interact with ThyA in vivo. The library also was screened for aptamers that inhibited growth of bacteria expressing them, and five such aptamers were characterized. Four aptamers were bacteriostatic when expressed, whereas one showed a bactericidal effect. Introduction of translational stop codons into various aptamers blocked their activity, suggesting that their biological effects were likely to be due to protein aptamer rather than RNA. Combinatorial aptamers provide a new genetic and biochemical tool for identifying targets for antibacterial drug development.
Collapse
Affiliation(s)
- J H Blum
- Department of Microbiology, Shipley Institute of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
33
|
Roland J, Berezov A, Greene MI, Murali R, Piatier-Tonneau D, Devaux C, Briant L. The synthetic CD4 exocyclic CDR3.AME(82-89) inhibits NF-kappaB nuclear translocation, HIV-1 promoter activation, and viral gene expression. DNA Cell Biol 1999; 18:819-28. [PMID: 10595395 DOI: 10.1089/104454999314818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have previously shown that the synthetic aromatically modified exocyclic (AME) analog (CDR3.AME(82-89), derived from the CDR3 (residues 82-89) region of CD4 domain 1, inhibits replication of human immunodeficiency virus type 1 (HIV-1) in infected cells. In this work, we investigated the mechanism by which this inhibition is achieved. Although cells exposed to HIV-1 and treated with the CDR3.AME(82-89) peptide did not release viral particles for more than a week and kept surface expression of CD4, viral DNA was found in those cells 24 h after virus exposure, indicating that the CDR3.AME(82-89) analog does not prevent virus entry. However, virus transcription remained extremely low in infected cells, as demonstrated by the study of spliced HIV-1 mRNA in cultures treated with CDR3.AME(82-89) 72 h postinfection. Finally, the CDR3.AME(82-89) peptide was found to be a potent inhibitor of HIV-1 promoter activity and nuclear factor-kappaB translocation, indicating that the antiviral property of this peptide is, at least in part, linked with the ability of the molecule to prevent HIV-1 transcription.
Collapse
Affiliation(s)
- J Roland
- Laboratoire Infections Rétrovirales et Signalisation Cellulaire, CNRS EP J0004, Institut de Biologie, Montpellier, France
| | | | | | | | | | | | | |
Collapse
|
34
|
Sakata K, Sakata A, Kong L, Vela-Roch N, Talal N, Dang H. Monocyte rescue of human T cells from apoptosis is CD40/CD154 dependent. Scand J Immunol 1999; 50:479-84. [PMID: 10564550 DOI: 10.1046/j.1365-3083.1999.00629.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The induction of T-cell apoptosis is regulated in part by monocytes (CD14+ cells). Human peripheral blood monocytes inhibited the spontaneous cell death of activated T cells in vitro. The inhibition of T-cell apoptosis did not require autologous monocytes. Inhibition required direct contact with monocytes and was not due to a soluble factor. Furthermore, treatment of monocytes with actinomycin D, cycloheximide and paraformaldehyde abrogated the anti-apoptotic activity of these cells. Blocking antibody to CD40 and CD154 (CD40 ligand) decreased the ability of monocytes to aid in T-cell survival, whereas, blocking LFA-1/I-CAM-1, Fas ligand and the CD4/major histocompatibility complex class II interaction did not affect the influence of monocytes on T-cell survival. This shows that monocytes rescue of activated T cells from apoptosis is dependent upon CD40/CD154 interaction.
Collapse
Affiliation(s)
- K Sakata
- University of Texas Health Science Center, San Antonio, TX 78284, USA
| | | | | | | | | | | |
Collapse
|
35
|
Major histocompatibility complex–derived peptides as novel forms of immunosuppression. Curr Opin Organ Transplant 1999. [DOI: 10.1097/00075200-199909000-00006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Kundu B, Khare SK. Recent advances in immunosuppressants. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 1999; 52:1-51. [PMID: 10396125 DOI: 10.1007/978-3-0348-8730-4_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
In recent years, a large number of structurally diverse immunosuppressants have been discovered that are effective for the treatment of organ transplantation. Some of them are undergoing clinical trials and may soon enter into routine clinical practice. These compounds are either chemical entities obtained from natural sources/synthetic means or biomaterials such as monoclonal antibodies/gene products/proteins. They have been found to interfere at different stages of T cell activation and proliferation, and can be identified as inhibitors of nucleotide synthesis, growth factor signal transduction and differentiation. Newer strategies involving combination of new agents with traditional immunosuppressants, monoclonal antibodies and gene therapy offer enormous potential, not only for the investigation of mechanisms pertaining to graft rejection, but also for its therapeutic prevention.
Collapse
Affiliation(s)
- B Kundu
- Division of Biopolymers, Central Drug Research Institute, Lucknow, India
| | | |
Collapse
|
37
|
Abstract
A growing body of experimental evidence demonstrates that synthetic peptides corresponding to linear sequences of MHC (HLA in humans) proteins have immunomodulatory effects in vitro and in vivo in animal models and in humans. Although the original concept was that these peptides inhibited antigen recognition at the MHC-T cell receptor interface via physical blockade, it is now clear that the mechanisms responsible for the myriad of functional effects are more complex. Recent findings show that some peptides affect signal transduction and cell cycle progression. Fragments of MHC molecules can dampen or downregulate immune responses via a variety of mechanisms. Some soluble MHC molecules or synthetic peptides are capable of inducing and maintaining immunologic tolerance in animals. This information suggests that synthetic peptides themselves or drugs mimicking their effects may represent a new class of immunotherapeutics.
Collapse
Affiliation(s)
- B Murphy
- Department of Medicine, Mount Sinai School of Medicine, New York, New York, USA
| | | |
Collapse
|
38
|
Kieber-Emmons T, Lin C, Foster MH, Kleyman TR. Antiidiotypic antibody recognizes an amiloride binding domain within the alpha subunit of the epithelial Na+ channel. J Biol Chem 1999; 274:9648-55. [PMID: 10092651 DOI: 10.1074/jbc.274.14.9648] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We previously raised an antibody (RA6.3) by an antiidiotypic approach which was designed to be directed against an amiloride binding domain on the epithelial Na+ channel (ENaC). This antibody mimicked amiloride in that it inhibited transepithelial Na+ transport across A6 cell monolayers. RA6.3 recognized a 72-kDa polypeptide in A6 epithelia treated with tunicamycin, consistent with the size of nonglycosylated Xenopus laevis alphaENaC. RA6.3 specifically recognized an amiloride binding domain within the alpha-subunit of mouse and bovine ENaC. The deduced amino acid sequence of RA6.3 was used to generate a three-dimensional model structure of the antibody. The combining site of RA6.3 was epitope mapped using a novel computer-based strategy. Organic residues that potentially interact with the RA6.3 combining site were identified by data base screening using the program LUDI. Selected residues docked to the antibody in a manner corresponding to the ordered linear array of amino acid residues within an amiloride binding domain on the alpha-subunit of ENaC. A synthetic peptide spanning this domain inhibited the binding of RA6.3 to alphaENaC. This analysis provided a novel approach to develop models of antibody-antigen interaction as well as a molecular perspective of RA6.3 binding to an amiloride binding domain within alphaENaC.
Collapse
Affiliation(s)
- T Kieber-Emmons
- Department of Pathology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
39
|
Deigin VI, Poverenny AM, Semina OV, Semenets TN. Reciprocal effect of optical isomerism of EW-dipeptides on immune response. Immunol Lett 1999; 67:41-6. [PMID: 10217204 DOI: 10.1016/s0165-2478(98)00149-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Dipeptides of the EW-sequence, consisting of D-amino acids, were shown to inhibit spleen colony formation both after in vitro treatment of bone marrow and after the peptide injection to the donor 2-48 h prior to bone marrow taking. The inhibiting effect of D-(EW) peptides can be eliminated by injection of thymic cells to the recipient. L-(EW) peptides have no influence on colony forming activity of intact bone marrow cells but stimulate regeneration of colony formation by irradiated (1 Gy) bone marrow. Unlike L-(EW), the D-(EW) peptides suppress the development of hemopoietic stem cells into the S-phase. Supposedly, D-(EW) represent a new generation of immunoactive peptides possessing of inhibiting activity.
Collapse
Affiliation(s)
- V I Deigin
- Peptide Engineering Centre PEPTOS, Miklukho-Maklaya, Moscow, Russian Federation.
| | | | | | | |
Collapse
|
40
|
Affiliation(s)
- C C Magee
- Laboratory of Immunogenetics and Transplantation, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
41
|
Li S, Choksi S, Shan S, Hu X, Gao J, Korngold R, Huang Z. Identification of the CD8 DE loop as a surface functional epitope. Implications for major histocompatibility complex class I binding and CD8 inhibitor design. J Biol Chem 1998; 273:16442-5. [PMID: 9632710 DOI: 10.1074/jbc.273.26.16442] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We used an approach of protein surface epitope mapping by synthetic peptides to analyze the surface structure-function relationship of the CD8 protein. Small synthetic peptide mimics of the CD8 DE loop were shown to effectively block CD8 binding to major histocompatibility complex (MHC) class I molecules and possess significant inhibitory activity on in vitro CD8(+) T cell function. These results suggested that the DE loop region of the CD8 protein is an important functional epitope mediating CD8-MHC class I interaction and the activation of CD8(+) T cells, a finding that is consistent with the recently reported crystal structure of the CD8-MHC class I complex. The structural basis for the biological activity of the DE loop peptide was further analyzed in a series of analogs containing alanine substitutions. This study provides support for the concept of bioactive peptide design based on protein surface epitopes and suggests that such an approach may be applicable to other protein-protein complexes, particularly those of immunoglobulin superfamily molecules.
Collapse
Affiliation(s)
- S Li
- Kimmel Cancer Institute, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Molecules involved in cell adhesion processes are often both structurally and functionally modular, with subdomains that are members of large protein families. Recently, high-resolution structures have been determined for representative members of many of these families including fragments of integrins, cadherins, fibronectin-like domains, and immunoglobulin-like domains. These structures have enhanced our understanding of cell adhesion processes at several levels. In almost all cases, ligand-binding sites have been visualized and provide insight into how these molecules mediate biologically important interactions. Metal-binding sites have been identified and characterized, allowing assessment of the role of bound ions in cell adhesion processes. Many of these structures serve as templates for modeling homologous domains in other proteins or, when the structure of a fragment consisting of more than one domain is determined, the structure of multidomain arrays of homologous domains. Knowledge of atomic structure also allows rational design of drugs that either mimic or target specific binding sites. In many cases, high-resolution structures have revealed unexpected relationships that pose questions about the evolutionary origin of specific domains. This review briefly describes several recently determined structures of cell adhesion molecules, summarizes some of the main results of each structure, and highlights common features of different systems.
Collapse
Affiliation(s)
- D J Leahy
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.
| |
Collapse
|
43
|
Affiliation(s)
- B A Helm
- Department of Molecular Biology, Krebs Institute for Biomolecular Research, University of Sheffield, UK
| | | | | |
Collapse
|
44
|
Abstract
New insights into the mechanisms of allorecognition and the interactions of the TCR with the MHC molecule-peptide complex on antigen presenting cells have focused attention on developing novel biological strategies to modify the alloimmune response. Peptides derived from various regions of MHC class I and II molecules and structure-based peptides have demonstrated immunomodulatory effects both in vitro and in vivo. Their binding sites and mechanisms of action are under active investigation. Trials in human transplant recipients, with an MHC class I peptide have already begun.
Collapse
Affiliation(s)
- C C Magee
- Laboratory of Immunogenetics and Transplantation, Brigham and Women's Hospital, Boston, MA, USA
| | | |
Collapse
|
45
|
Satoh T, Aramini JM, Li S, Friedman TM, Gao J, Edling AE, Townsend R, Koch U, Choksi S, Germann MW, Korngold R, Huang Z. Bioactive peptide design based on protein surface epitopes. A cyclic heptapeptide mimics CD4 domain 1 CC' loop and inhibits CD4 biological function. J Biol Chem 1997; 272:12175-80. [PMID: 9115290 DOI: 10.1074/jbc.272.18.12175] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The interaction between CD4 and major histocompatibility complex class II proteins provides a critical co-receptor function for the activation of CD4(+) T cells implicated in the pathogenesis of a number of autoimmune diseases and transplantation responses. A small synthetic cyclic heptapeptide was designed and shown by high resolution NMR spectroscopy to closely mimic the CD4 domain 1 CC' surface loop. This peptide effectively blocked stable CD4-major histocompatibility complex class II interaction, possessed significant immunosuppressive activity in vitro and in vivo, and strongly resisted proteolytic degradation. These results demonstrate the therapeutic potential of this peptide as a novel immunosuppressive agent and suggest a general strategy of drug design by using small conformationally constrained peptide mimics of protein surface epitopes to inhibit protein interactions and biological functions.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Binding Sites
- CD4 Antigens/chemistry
- CD4 Antigens/drug effects
- Cell Adhesion/drug effects
- Computer Graphics
- Drug Design
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Epitopes/chemistry
- Female
- Genes, MHC Class II
- Graft Rejection/immunology
- Graft Survival/drug effects
- Graft vs Host Disease
- Humans
- Lymphocyte Culture Test, Mixed
- Lymphocytes/immunology
- Lymphocytes/radiation effects
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Models, Molecular
- Oligopeptides/chemical synthesis
- Oligopeptides/chemistry
- Oligopeptides/pharmacology
- Peptides, Cyclic/chemical synthesis
- Peptides, Cyclic/chemistry
- Protein Structure, Secondary
- Skin Transplantation/immunology
Collapse
Affiliation(s)
- T Satoh
- Kimmel Cancer Institute, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|