1
|
Loharch S, Medina-Trillo C, Sedgwick DM, Barrio P, Fustero S, Gallego J. Bilaterally Substituted Terphenyl Molecules Efficiently Inhibit the Interaction between a Protein and a Fully Buried α-Helix in the Malaria Parasite Motor System. J Am Chem Soc 2025; 147:15917-15922. [PMID: 40316890 DOI: 10.1021/jacs.4c15031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Abstract
Protein-protein interactions (PPI) frequently involve α-helices and are challenging targets for small-molecule drugs. Here we report the design, synthesis and evaluation of new PPI inhibitors based on a bilaterally substituted p-terphenyl scaffold. The side groups of this scaffold are projected in a broad spatial angle and reproduced the interactions of the myosin A (MyoA) α-helix wrapped by the Myosin Tail Interacting Protein (MTIP) in Plasmodium parasites causing malaria. Fluorescence, calorimetry, and NMR spectroscopy analyses revealed that the terphenyl molecules recognized the MyoA binding site within the MTIP and were capable of displacing the α-helix from its protein receptor and triggering comparable conformational changes in MTIP. The MTIP affinity of the best inhibitor was strikingly close to that exhibited by the MyoA helix. These data indicate that a small-molecule terphenyl compound can efficiently mimic a four-times heavier polypeptide. These molecules may serve as probes for PPIs involving deeply buried α-helices.
Collapse
Affiliation(s)
- Saurabh Loharch
- Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia, C/Quevedo 2, 46001 Valencia, Spain
| | - Cristina Medina-Trillo
- Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia, C/Quevedo 2, 46001 Valencia, Spain
| | - Daniel M Sedgwick
- Universidad de Valencia, Avda. V. A. Estellés s/n, 46100 Burjassot, Spain
| | - Pablo Barrio
- Universidad de Valencia, Avda. V. A. Estellés s/n, 46100 Burjassot, Spain
| | - Santos Fustero
- Universidad de Valencia, Avda. V. A. Estellés s/n, 46100 Burjassot, Spain
| | - José Gallego
- Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia, C/Quevedo 2, 46001 Valencia, Spain
| |
Collapse
|
2
|
Waterworth SC, Shenoy SR, Sharma ND, Wolcott C, Donohue DE, O'Keefe BR, Beutler JA. ShiftScan: A tool for rapid analysis of high-throughput differential scanning fluorimetry data and compound prioritization. Protein Sci 2025; 34:e70055. [PMID: 39989223 PMCID: PMC11848206 DOI: 10.1002/pro.70055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/25/2025]
Abstract
Differential scanning fluorimetry (DSF) can be an effective high-throughput screening assay in drug discovery for detecting protein-compound interactions that stabilize or destabilize macromolecules. Due to the magnitude and quality of the data produced by this biophysical assay, analyzing and prioritizing compounds from large-scale DSF data sets has proven challenging to the research community. Here, we present ShiftScan-a powerful, stand-alone tool designed for the rapid analysis of DSF data and compound prioritization based on thermal transition patterns. ShiftScan accurately and quickly predicts melting temperatures (Tm values) from both canonical and non-canonical transition patterns, efficiently filtering out spurious data to minimize false positives. We report on the use of this tool for data analysis of screens involving both pure compound and natural product fraction libraries and provide the software to the screening community to aid in the discovery of molecularly-targeted compounds. Instructions for installation and usage of ShiftScan can be found at our GitHub repository: https://github.com/samche42/ShiftScan.
Collapse
Affiliation(s)
- Samantha C. Waterworth
- Molecular Targets Program, Center for Cancer ResearchNational Cancer InstituteFrederickMarylandUSA
| | - Shilpa R. Shenoy
- Molecular Targets Program, Center for Cancer ResearchNational Cancer InstituteFrederickMarylandUSA
| | - Nirmala D. Sharma
- Molecular Targets Program, Center for Cancer ResearchNational Cancer InstituteFrederickMarylandUSA
| | - Chris Wolcott
- Molecular Targets Program, Center for Cancer ResearchNational Cancer InstituteFrederickMarylandUSA
- Advanced Biomedical Computational ScienceFrederick National Laboratory for Cancer ResearchFrederickMarylandUSA
| | - Duncan E. Donohue
- Statistics Department, Data Management Services Inc.Frederick National Laboratory for Cancer Research Sponsored by the National Cancer InstituteFrederickMarylandUSA
| | - Barry R. O'Keefe
- Molecular Targets Program, Center for Cancer ResearchNational Cancer InstituteFrederickMarylandUSA
- Natural Products Branch, Developmental Therapeutics Program, Division of Cancer Treatment and DiagnosisNational Cancer InstituteFrederickMarylandUSA
| | - John A. Beutler
- Molecular Targets Program, Center for Cancer ResearchNational Cancer InstituteFrederickMarylandUSA
| |
Collapse
|
3
|
Giannakou M, Akrani I, Tsoka A, Myrianthopoulos V, Mikros E, Vorgias C, Hatzinikolaou DG. Discovery of Novel Inhibitors against ALS-Related SOD1(A4V) Aggregation through the Screening of a Chemical Library Using Differential Scanning Fluorimetry (DSF). Pharmaceuticals (Basel) 2024; 17:1286. [PMID: 39458929 PMCID: PMC11510448 DOI: 10.3390/ph17101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Cu/Zn Superoxide Dismutase 1 (SOD1) is a 32 kDa cytosolic dimeric metalloenzyme that neutralizes superoxide anions into oxygen and hydrogen peroxide. Mutations in SOD1 are associated with ALS, a disease causing motor neuron atrophy and subsequent mortality. These mutations exert their harmful effects through a gain of function mechanism, rather than a loss of function. Despite extensive research, the mechanism causing selective motor neuron death still remains unclear. A defining feature of ALS pathogenesis is protein misfolding and aggregation, evidenced by ubiquitinated protein inclusions containing SOD1 in affected motor neurons. This work aims to identify compounds countering SOD1(A4V) misfolding and aggregation, which could potentially aid in ALS treatment. METHODS The approach employed was in vitro screening of a library comprising 1280 pharmacologically active compounds (LOPAC®) in the context of drug repurposing. Using differential scanning fluorimetry (DSF), these compounds were tested for their impact on SOD1(A4V) thermal stability. RESULTS AND CONCLUSIONS Dimer stability was the parameter chosen as the criterion for screening, since the dissociation of the native SOD1 dimer is the step prior to its in vitro aggregation. The screening revealed one compound raising protein-ligand Tm by 6 °C, eleven inducing a higher second Tm, suggesting a stabilization effect, and fourteen reducing Tm from 10 up to 26 °C, suggesting possible interactions or non-specific binding.
Collapse
Affiliation(s)
- Maria Giannakou
- Biochemistry and Molecular Biology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
- Enzyme and Microbial Biotechnology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| | - Ifigeneia Akrani
- Laboratory of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15784 Zografou, Greece; (I.A.)
| | - Angeliki Tsoka
- Biochemistry and Molecular Biology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| | - Vassilios Myrianthopoulos
- Laboratory of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15784 Zografou, Greece; (I.A.)
| | - Emmanuel Mikros
- Laboratory of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15784 Zografou, Greece; (I.A.)
| | - Constantinos Vorgias
- Biochemistry and Molecular Biology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| | - Dimitris G. Hatzinikolaou
- Enzyme and Microbial Biotechnology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| |
Collapse
|
4
|
Kelley EH, Minasov G, Konczak K, Shuvalova L, Brunzelle JS, Shukla S, Beulke M, Thabthimthong T, Olsen KW, Inniss NL, Satchell KJF, Becker DP. Biochemical and Structural Analysis of the Bacterial Enzyme Succinyl-Diaminopimelate Desuccinylase (DapE) from Acinetobacter baumannii. ACS OMEGA 2024; 9:3905-3915. [PMID: 38284080 PMCID: PMC10809365 DOI: 10.1021/acsomega.3c08231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 01/30/2024]
Abstract
There is an urgent need for new antibiotics given the rise of antibiotic resistance, and succinyl-diaminopimelate desuccinylase (DapE, E.C. 3.5.1.18) has emerged as a promising bacterial enzyme target. DapE from Haemophilus influenzae (HiDapE) has been studied and inhibitors identified, but it is essential to explore DapE from different species to assess selective versus broad-spectrum therapeutics. We have determined the structure of DapE from the ESKAPE pathogen Acinetobacter baumannii (AbDapE) and studied inhibition by known inhibitors of HiDapE. AbDapE is inhibited by captopril and sulfate comparable to HiDapE, but AbDapE was not significantly inhibited by a known indoline sulfonamide HiDapE inhibitor. Captopril and sulfate both stabilize HiDapE by increasing the thermal melting temperature (Tm) in thermal shift assays. By contrast, sulfate decreases the stability of the AbDapE enzyme, whereas captopril increases the stability. Further, we report two crystal structures of selenomethionine-substituted AbDapE in the closed conformation, one with AbDapE in complex with succinate derived from enzymatic hydrolysis of N6-methyl-l,l-SDAP substrate and acetate (PDB code 7T1Q, 2.25 Å resolution), and a crystal structure of AbDapE with bound succinate along with l-(S)-lactate, a product of degradation of citric acid from the crystallization buffer during X-ray irradiation (PDB code 8F8O, 2.10 Å resolution).
Collapse
Affiliation(s)
- Emma H. Kelley
- Department
of Chemistry and Biochemistry, Loyola University
Chicago, 1032 West Sheridan Road, Chicago, Illinois 60660, United States
| | - George Minasov
- Department
of Microbiology-Immunology, Northwestern
University, Feinberg School of Medicine, Chicago, Illinois 60611, United States
- Center
for Structural Biology of Infectious Diseases, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Katherine Konczak
- Department
of Chemistry and Biochemistry, Loyola University
Chicago, 1032 West Sheridan Road, Chicago, Illinois 60660, United States
| | - Ludmilla Shuvalova
- Department
of Pharmacology, Northwestern University,
Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Joseph S. Brunzelle
- Northwestern
Synchrotron Research Center, Life Sciences Collaborative Access Team, Northwestern University, Argonne, Illinois 60439, United States
| | - Shantanu Shukla
- Department
of Microbiology-Immunology, Northwestern
University, Feinberg School of Medicine, Chicago, Illinois 60611, United States
- Center
for Structural Biology of Infectious Diseases, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Megan Beulke
- Department
of Chemistry and Biochemistry, Loyola University
Chicago, 1032 West Sheridan Road, Chicago, Illinois 60660, United States
| | - Teerana Thabthimthong
- Department
of Chemistry and Biochemistry, Loyola University
Chicago, 1032 West Sheridan Road, Chicago, Illinois 60660, United States
| | - Kenneth W. Olsen
- Department
of Chemistry and Biochemistry, Loyola University
Chicago, 1032 West Sheridan Road, Chicago, Illinois 60660, United States
| | - Nicole L. Inniss
- Department
of Microbiology-Immunology, Northwestern
University, Feinberg School of Medicine, Chicago, Illinois 60611, United States
- Center
for Structural Biology of Infectious Diseases, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Karla J. F. Satchell
- Department
of Microbiology-Immunology, Northwestern
University, Feinberg School of Medicine, Chicago, Illinois 60611, United States
- Center
for Structural Biology of Infectious Diseases, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Daniel P. Becker
- Department
of Chemistry and Biochemistry, Loyola University
Chicago, 1032 West Sheridan Road, Chicago, Illinois 60660, United States
| |
Collapse
|
5
|
Habeeb Mohammad TS, Kelley EH, Reidl CT, Konczak K, Beulke M, Javier J, Olsen KW, Becker DP. Cyclobutanone Inhibitors of Diaminopimelate Desuccinylase (DapE) as Potential New Antibiotics. Int J Mol Sci 2024; 25:1339. [PMID: 38279338 PMCID: PMC10815964 DOI: 10.3390/ijms25021339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Based on our previous success in using cyclobutanone derivatives as enzyme inhibitors, we have designed and prepared a 37-member library of α-aminocyclobutanone amides and sulfonamides, screened for inhibition of the bacterial enzyme diaminopimelate desuccinylase (DapE), which is a promising antibiotic target, and identified several inhibitors with micromolar inhibitory potency. Molecular docking suggests binding of the deprotonated hydrate of the strained cyclobutanone, and thermal shift analysis with the most potent inhibitor (3y, IC50 = 23.1 µM) enabled determination of a Ki value of 10.2 +/- 0.26 µM and observed two separate Tm values for H. influenzae DapE (HiDapE).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Daniel P. Becker
- Department of Chemistry and Biochemistry, Loyola University Chicago, 1032 West Sheridan Road, Chicago, IL 60660, USA; (T.S.H.M.); (E.H.K.); (K.K.); (M.B.); (J.J.); (K.W.O.)
| |
Collapse
|
6
|
He L, Qiu Y, Pang G, Li S, Wang J, Feng Y, Chen L, Zhu L, Liu Y, Cui L, Cao Y, Zhu X. Plasmodium falciparum GAP40 Plays an Essential Role in Merozoite Invasion and Gametocytogenesis. Microbiol Spectr 2023; 11:e0143423. [PMID: 37249423 PMCID: PMC10269477 DOI: 10.1128/spectrum.01434-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
Cyclic invasion of red blood cells (RBCs) by Plasmodium merozoites is associated with the symptoms and pathology of malaria. Merozoite invasion is powered actively and rapidly by a parasite actomyosin motor called the glideosome. The ability of the glideosome to generate force to support merozoite entry into the host RBCs is thought to rely on its stable anchoring within the inner membrane complex (IMC) through membrane-resident proteins, such as GAP50 and GAP40. Using a conditional knockdown (KD) approach, we determined that PfGAP40 was required for asexual blood-stage replication. PfGAP40 is not needed for merozoite egress from host RBCs or for the attachment of merozoites to new RBCs. PfGAP40 coprecipitates with PfGAP45 and PfGAP50. During merozoite invasion, PfGAP40 is associated strongly with stabilizing the expression levels of PfGAP45 and PfGAP50 in the schizont stage. Although PfGAP40 KD did not influence IMC integrity, it impaired the maturation of gametocytes. In addition, PfGAP40 is phosphorylated, and mutations that block phosphorylation of PfGAP40 at the C-terminal serine residues S370, S372, S376, S405, S409, S420, and S445 reduced merozoite invasion efficiency. Overall, our findings implicate PfGAP40 as an important regulator for the gliding activity of merozoites and suggest that phosphorylation is required for PfGAP40 function. IMPORTANCE Red blood cell invasion is central to the pathogenesis of the malaria parasite, and the parasite proteins involved in this process are potential therapeutic targets. Gliding motility powers merozoite invasion and is driven by a unique molecular motor termed the glideosome. The glideosome is stably anchored to the parasite inner membrane complex (IMC) through membrane-resident proteins. In the present study, we demonstrate the importance of an IMC-resident glideosome component, PfGAP40, that plays a critical role in stabilizing the expression levels of glideosome components in the schizont stage. We determined that phosphorylation of PfGAP40 at C-terminal residues is required for efficient merozoite invasion.
Collapse
Affiliation(s)
- Lu He
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Yue Qiu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Geping Pang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Siqi Li
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Jingjing Wang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Yonghui Feng
- Department of Laboratory Medicine, the First Hospital of China Medical University, Shenyang, Liaoning, China
- National Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning, China
| | - Lumeng Chen
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Liying Zhu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Yinjie Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Liwang Cui
- College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Xiaotong Zhu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
7
|
Terrazas-López M, Lobo-Galo N, Aguirre-Reyes LG, Bustos-Jaimes I, Marcos-Víquez JÁ, González-Segura L, Díaz-Sánchez ÁG. Interaction of N-succinyl diaminopimelate desuccinylase with orphenadrine and disulfiram. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
8
|
Anam ZE, Joshi N, Gupta S, Yadav P, Chaurasiya A, Kahlon AK, Kaushik S, Munde M, Ranganathan A, Singh S. A De novo Peptide from a High Throughput Peptide Library Blocks Myosin A -MTIP Complex Formation in Plasmodium falciparum. Int J Mol Sci 2020; 21:ijms21176158. [PMID: 32859024 PMCID: PMC7503848 DOI: 10.3390/ijms21176158] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/09/2020] [Accepted: 05/15/2020] [Indexed: 01/09/2023] Open
Abstract
Apicomplexan parasites, through their motor machinery, produce the required propulsive force critical for host cell-entry. The conserved components of this so-called glideosome machinery are myosin A and myosin A Tail Interacting Protein (MTIP). MTIP tethers myosin A to the inner membrane complex of the parasite through 20 amino acid-long C-terminal end of myosin A that makes direct contacts with MTIP, allowing the invasion of Plasmodium falciparum in erythrocytes. Here, we discovered through screening a peptide library, a de-novo peptide ZA1 that binds the myosin A tail domain. We demonstrated that ZA1 bound strongly to myosin A tail and was able to disrupt the native myosin A tail MTIP complex both in vitro and in vivo. We then showed that a shortened peptide derived from ZA1, named ZA1S, was able to bind myosin A and block parasite invasion. Overall, our study identified a novel anti-malarial peptide that could be used in combination with other antimalarials for blocking the invasion of Plasmodium falciparum.
Collapse
Affiliation(s)
- Zill e Anam
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Nishant Joshi
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, Uttar Pradesh 201304, India;
| | - Sakshi Gupta
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India; (S.G.); (M.M.)
| | - Preeti Yadav
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Ayushi Chaurasiya
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Amandeep Kaur Kahlon
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Shikha Kaushik
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Manoj Munde
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India; (S.G.); (M.M.)
| | - Anand Ranganathan
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
- Correspondence: (A.R.); (S.S.)
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
- Correspondence: (A.R.); (S.S.)
| |
Collapse
|
9
|
Gao K, Oerlemans R, Groves MR. Theory and applications of differential scanning fluorimetry in early-stage drug discovery. Biophys Rev 2020; 12:85-104. [PMID: 32006251 PMCID: PMC7040159 DOI: 10.1007/s12551-020-00619-2] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/08/2020] [Indexed: 02/06/2023] Open
Abstract
Differential scanning fluorimetry (DSF) is an accessible, rapid, and economical biophysical technique that has seen many applications over the years, ranging from protein folding state detection to the identification of ligands that bind to the target protein. In this review, we discuss the theory, applications, and limitations of DSF, including the latest applications of DSF by ourselves and other researchers. We show that DSF is a powerful high-throughput tool in early drug discovery efforts. We place DSF in the context of other biophysical methods frequently used in drug discovery and highlight their benefits and downsides. We illustrate the uses of DSF in protein buffer optimization for stability, refolding, and crystallization purposes and provide several examples of each. We also show the use of DSF in a more downstream application, where it is used as an in vivo validation tool of ligand-target interaction in cell assays. Although DSF is a potent tool in buffer optimization and large chemical library screens when it comes to ligand-binding validation and optimization, orthogonal techniques are recommended as DSF is prone to false positives and negatives.
Collapse
Affiliation(s)
- Kai Gao
- Structure Biology in Drug Design, Drug Design Group XB20, Departments of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Rick Oerlemans
- Structure Biology in Drug Design, Drug Design Group XB20, Departments of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Matthew R Groves
- Structure Biology in Drug Design, Drug Design Group XB20, Departments of Pharmacy, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
10
|
Wall RJ, Zeeshan M, Katris NJ, Limenitakis R, Rea E, Stock J, Brady D, Waller RF, Holder AA, Tewari R. Systematic analysis of Plasmodium myosins reveals differential expression, localisation, and function in invasive and proliferative parasite stages. Cell Microbiol 2019; 21:e13082. [PMID: 31283102 PMCID: PMC6851706 DOI: 10.1111/cmi.13082] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/13/2019] [Accepted: 07/03/2019] [Indexed: 11/28/2022]
Abstract
The myosin superfamily comprises of actin-dependent eukaryotic molecular motors important in a variety of cellular functions. Although well studied in many systems, knowledge of their functions in Plasmodium, the causative agent of malaria, is restricted. Previously, six myosins were identified in this genus, including three Class XIV myosins found only in Apicomplexa and some Ciliates. The well characterized MyoA is a Class XIV myosin essential for gliding motility and invasion. Here, we characterize all other Plasmodium myosins throughout the parasite life cycle and show that they have very diverse patterns of expression and cellular location. MyoB and MyoE, the other two Class XIV myosins, are expressed in all invasive stages, with apical and basal locations, respectively. Gene deletion revealed that MyoE is involved in sporozoite traversal, MyoF and MyoK are likely essential in the asexual blood stages, and MyoJ and MyoB are not essential. Both MyoB and its essential light chain (MCL-B) are localised at the apical end of ookinetes but expressed at completely different time points. This work provides a better understanding of the role of actomyosin motors in Apicomplexan parasites, particularly in the motile and invasive stages of Plasmodium during sexual and asexual development within the mosquito.
Collapse
Affiliation(s)
- Richard J. Wall
- School of Life Sciences, Queens Medical CentreUniversity of NottinghamNottinghamUK
| | - Mohammad Zeeshan
- School of Life Sciences, Queens Medical CentreUniversity of NottinghamNottinghamUK
| | | | | | - Edward Rea
- School of Life Sciences, Queens Medical CentreUniversity of NottinghamNottinghamUK
| | - Jessica Stock
- School of Life Sciences, Queens Medical CentreUniversity of NottinghamNottinghamUK
| | - Declan Brady
- School of Life Sciences, Queens Medical CentreUniversity of NottinghamNottinghamUK
| | - Ross F. Waller
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | | | - Rita Tewari
- School of Life Sciences, Queens Medical CentreUniversity of NottinghamNottinghamUK
| |
Collapse
|
11
|
In vitro interaction between Plasmodium falciparum myosin B (PfMyoB) and myosin A tail interacting protein (MTIP). Parasitol Res 2018; 117:3437-3446. [PMID: 30094538 DOI: 10.1007/s00436-018-6039-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/02/2018] [Indexed: 10/28/2022]
Abstract
Apicomplexan parasites, including Plasmodium falciparum, are obligate intracellular organisms that utilize a strategy termed "gliding" to move and invade host cells, causing disease. Gliding is carried out by a protein complex known as the glideosome, which includes an actin-myosin motor. To date, six myosins have been identified in P. falciparum (PfMyoA, B, C, D, E, and F), but only the role of PfMyoA, the myosin of the glideosome that is involved in the process of red blood cell and mosquito cell invasion, has been established. Based on previous observations, we speculated that PfMyoA and PfMyoB may have similar or redundant functions. To test this hypothesis, we searched for in vitro interactions between PfMyoB and MTIP (myosin A tail interacting protein), the myosin light chain of PfMyoA. A set of differentially tagged PfMyoA, PfMyoB, and MTIP recombinant proteins was employed to specifically and simultaneously detect each myosin in competition assays and inhibition assays using specific peptides. MTIP potentially acts as the light chain of PfMyoB.
Collapse
|
12
|
Boucher LE, Hopp CS, Muthinja JM, Frischknecht F, Bosch J. Discovery of Plasmodium (M)TRAP-Aldolase Interaction Stabilizers Interfering with Sporozoite Motility and Invasion. ACS Infect Dis 2018; 4:620-634. [PMID: 29411968 DOI: 10.1021/acsinfecdis.7b00225] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As obligate, intracellular parasites, Plasmodium spp. rely on invasion of host cells in order to replicate and continue their life cycle. The parasite needs to traverse the dermis and endothelium of blood vessels, invade hepatocytes and red blood cells, traverse the mosquito midgut, and enter the salivary glands to continue the cycle of infection and transmission. To traverse and invade cells, the parasite employs an actomyosin motor at the core of a larger invasion machinery complex known as the glideosome. The complex is comprised of multiple protein-protein interactions linking the motor to the internal cytoskeletal network of the parasite and to the extracellular adhesins, which directly contact the host tissue or cell surface. One key interaction is between the cytoplasmic tails of the thrombospondin related anonymous protein (TRAP) and aldolase, a bridging protein to the motor. Here, we present results from screening the Medicines for Malaria Venture (MMV) library of 400 compounds against this key protein-protein interaction. Using a surface plasmon resonance screen, we have identified several compounds that modulate the dynamics of the interaction between TRAP and aldolase. These compounds have been validated in vitro by studying their effects on sporozoite gliding motility and hepatocyte invasion. One of the MMV compounds identified reduced invasion levels by 89% at the lowest concentration tested (16 μM) and severely inhibited gliding at even lower concentrations (5 μM). By targeting protein-protein interactions, we investigated an under-explored area of parasite biology and general drug development, to identify potential antimalarial lead compounds.
Collapse
Affiliation(s)
- Lauren E. Boucher
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Christine S. Hopp
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Lane, Rockville, Maryland 20852, United States
| | - Julianne Mendi Muthinja
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Jürgen Bosch
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, Maryland 21205, United States
- InterRayBio, LLC, 1812 Ashland Avenue, Baltimore, Maryland 21205, United States
| |
Collapse
|
13
|
Powell CJ, Jenkins ML, Parker ML, Ramaswamy R, Kelsen A, Warshaw DM, Ward GE, Burke JE, Boulanger MJ. Dissecting the molecular assembly of the Toxoplasma gondii MyoA motility complex. J Biol Chem 2017; 292:19469-19477. [PMID: 28972141 DOI: 10.1074/jbc.m117.809632] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/22/2017] [Indexed: 01/28/2023] Open
Abstract
Apicomplexan parasites such as Toxoplasma gondii rely on a unique form of locomotion known as gliding motility. Generating the mechanical forces to support motility are divergent class XIV myosins (MyoA) coordinated by accessory proteins known as light chains. Although the importance of the MyoA-light chain complex is well-established, the detailed mechanisms governing its assembly and regulation are relatively unknown. To establish a molecular blueprint of this dynamic complex, we first mapped the adjacent binding sites of light chains MLC1 and ELC1 on the MyoA neck (residues 775-818) using a combination of hydrogen-deuterium exchange mass spectrometry and isothermal titration calorimetry. We then determined the 1.85 Å resolution crystal structure of MLC1 in complex with its cognate MyoA peptide. Structural analysis revealed a bilobed architecture with MLC1 clamping tightly around the helical MyoA peptide, consistent with the stable 10 nm Kd measured by isothermal titration calorimetry. We next showed that coordination of calcium by an EF-hand in ELC1 and prebinding of MLC1 to the MyoA neck enhanced the affinity of ELC1 for the MyoA neck 7- and 8-fold, respectively. When combined, these factors enhanced ELC1 binding 49-fold (to a Kd of 12 nm). Using the full-length MyoA motor (residues 1-831), we then showed that, in addition to coordinating the neck region, ELC1 appears to engage the MyoA converter subdomain, which couples the motor domain to the neck. These data support an assembly model where staged binding events cooperate to yield high-affinity complexes that are able to maximize force transduction.
Collapse
Affiliation(s)
- Cameron J Powell
- From the Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada and
| | - Meredith L Jenkins
- From the Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada and
| | - Michelle L Parker
- From the Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada and
| | - Raghavendran Ramaswamy
- From the Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada and
| | - Anne Kelsen
- the Departments of Microbiology and Molecular Genetics and
| | - David M Warshaw
- Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont 05405
| | - Gary E Ward
- the Departments of Microbiology and Molecular Genetics and
| | - John E Burke
- From the Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada and
| | - Martin J Boulanger
- From the Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada and
| |
Collapse
|
14
|
Myosin B of Plasmodium falciparum (PfMyoB): in silico prediction of its three-dimensional structure and its possible interaction with MTIP. Parasitol Res 2017; 116:1373-1382. [PMID: 28265752 DOI: 10.1007/s00436-017-5417-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 02/21/2017] [Indexed: 10/24/2022]
Abstract
The mobility and invasion strategy of Plasmodium falciparum is governed by a protein complex known as the glideosome, which contains an actin-myosin motor. It has been shown that myosin A of the parasite (PfMyoA) is the myosin of the glideosome, and the interaction of PfMyoA with myosin tail domain interacting protein (MTIP) determines its correct location and its ability to function in the complex. Because PfMyoA and myosin B of P. falciparum (PfMyoB) share high sequence identity, are both small proteins without a tail domain, belong to the class XIV myosins, and are expressed in late schizonts and merozoites, we suspect that these myosins may have similar or redundant functions. Therefore, this work examined the structural similarity between PfMyoA and PfMyoB and performed a molecular docking between PfMyoB and MTIP. Three-dimensional (3D) models obtained for PfMyoA and PfMyoB achieved high scores in the structural validation programs used, and their superimposition revealed high structural similarity, supporting the hypothesis of possible similar functions for these two proteins. The 3D interaction models obtained and energy values found suggested that interaction between PfMyoB and MTIP is possible. Given the apparent abundance of PfMyoA relative to PfMyoB in the parasite, we believe that the interaction between PfMyoB and MTIP would only be detectable in specific cellular environments because under normal circumstances, it would be masked by the interaction between PfMyoA and MTIP.
Collapse
|
15
|
Abstract
INTRODUCTION Despite the fact that diseases caused by protozoan parasites represent serious challenges for public health, animal production and welfare, only a limited panel of drugs has been marketed for clinical applications. AREAS COVERED Herein, the authors investigate two strategies, namely whole organism screening and target-based drug design. The present pharmacopoeia has resulted from whole organism screening, and the mode of action and targets of selected drugs are discussed. However, the more recent extensive genome sequencing efforts and the development of dry and wet lab genomics and proteomics that allow high-throughput screening of interactions between micromolecules and recombinant proteins has resulted in target-based drug design as the predominant focus in anti-parasitic drug development. Selected examples of target-based drug design studies are presented, and calcium-dependent protein kinases, important drug targets in apicomplexan parasites, are discussed in more detail. EXPERT OPINION Despite the enormous efforts in target-based drug development, this approach has not yet generated market-ready antiprotozoal drugs. However, whole-organism screening approaches, comprising of both in vitro and in vivo investigations, should not be disregarded. The repurposing of already approved and marketed drugs could be a suitable strategy to avoid fastidious approval procedures, especially in the case of neglected or veterinary parasitoses.
Collapse
Affiliation(s)
- Joachim Müller
- a Institute of Parasitology, Vetsuisse Faculty , University of Bern , Bern , Switzerland
| | - Andrew Hemphill
- a Institute of Parasitology, Vetsuisse Faculty , University of Bern , Bern , Switzerland
| |
Collapse
|
16
|
Winter A, Sigurdardottir AG, DiCara D, Valenti G, Blundell TL, Gherardi E. Developing Antagonists for the Met-HGF/SF Protein–Protein Interaction Using a Fragment-Based Approach. Mol Cancer Ther 2015; 15:3-14. [DOI: 10.1158/1535-7163.mct-15-0446] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/29/2015] [Indexed: 11/16/2022]
|
17
|
Nemetski SM, Cardozo TJ, Bosch G, Weltzer R, O'Malley K, Ejigiri I, Kumar KA, Buscaglia CA, Nussenzweig V, Sinnis P, Levitskaya J, Bosch J. Inhibition by stabilization: targeting the Plasmodium falciparum aldolase-TRAP complex. Malar J 2015; 14:324. [PMID: 26289816 PMCID: PMC4545932 DOI: 10.1186/s12936-015-0834-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/02/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Emerging resistance of the malaria parasite Plasmodium to current therapies underscores the critical importance of exploring novel strategies for disease eradication. Plasmodium species are obligate intracellular protozoan parasites. They rely on an unusual form of substrate-dependent motility for their migration on and across host-cell membranes and for host cell invasion. This peculiar motility mechanism is driven by the 'glideosome', an actin-myosin associated, macromolecular complex anchored to the inner membrane complex of the parasite. Myosin A, actin, aldolase, and thrombospondin-related anonymous protein (TRAP) constitute the molecular core of the glideosome in the sporozoite, the mosquito stage that brings the infection into mammals. METHODS Virtual library screening of a large compound library against the PfAldolase-TRAP complex was used to identify candidate compounds that stabilize and prevent the disassembly of the glideosome. The mechanism of these compounds was confirmed by biochemical, biophysical and parasitological methods. RESULTS A novel inhibitory effect on the parasite was achieved by stabilizing a protein-protein interaction within the glideosome components. Compound 24 disrupts the gliding and invasive capabilities of Plasmodium parasites in in vitro parasite assays. A high-resolution, ternary X-ray crystal structure of PfAldolase-TRAP in complex with compound 24 confirms the mode of interaction and serves as a platform for future ligand optimization. CONCLUSION This proof-of-concept study presents a novel approach to anti-malarial drug discovery and design. By strengthening a protein-protein interaction within the parasite, an avenue towards inhibiting a previously "undruggable" target is revealed and the motility motor responsible for successful invasion of host cells is rendered inactive. This study provides new insights into the malaria parasite cell invasion machinery and convincingly demonstrates that liver cell invasion is dramatically reduced by 95 % in the presence of the small molecule stabilizer compound 24.
Collapse
Affiliation(s)
- Sondra Maureen Nemetski
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, USA. .,Department of Pediatrics, Phyllis and David Komansky Center for Children's Health, New York-Presbyterian Hospital-Weill Cornell Medical College, New York, USA.
| | - Timothy J Cardozo
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, USA. .,Institute for Systems Genetics, New York University School of Medicine, New York, USA.
| | - Gundula Bosch
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA. .,Johns Hopkins Malaria Research Institute (JHMRI), Baltimore, USA.
| | - Ryan Weltzer
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA. .,Johns Hopkins Malaria Research Institute (JHMRI), Baltimore, USA.
| | - Kevin O'Malley
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA. .,Johns Hopkins Malaria Research Institute (JHMRI), Baltimore, USA.
| | - Ijeoma Ejigiri
- Department of Medical Parasitology, New York University School of Medicine, New York, USA.
| | - Kota Arun Kumar
- Michael Heidelberg Division of Pathology of Infectious Diseases, Department of Pathology, New York University School of Medicine, New York, USA. .,Department of Animal Sciences, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India.
| | - Carlos A Buscaglia
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de General San Martín-CONICET, 1650, San Martín, Buenos Aires, Argentina.
| | - Victor Nussenzweig
- Michael Heidelberg Division of Pathology of Infectious Diseases, Department of Pathology, New York University School of Medicine, New York, USA.
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA. .,Department of Medical Parasitology, New York University School of Medicine, New York, USA. .,Johns Hopkins Malaria Research Institute (JHMRI), Baltimore, USA.
| | - Jelena Levitskaya
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA. .,Johns Hopkins Malaria Research Institute (JHMRI), Baltimore, USA.
| | - Jürgen Bosch
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA. .,Johns Hopkins Malaria Research Institute (JHMRI), Baltimore, USA.
| |
Collapse
|
18
|
Laraia L, McKenzie G, Spring DR, Venkitaraman AR, Huggins DJ. Overcoming Chemical, Biological, and Computational Challenges in the Development of Inhibitors Targeting Protein-Protein Interactions. CHEMISTRY & BIOLOGY 2015; 22:689-703. [PMID: 26091166 PMCID: PMC4518475 DOI: 10.1016/j.chembiol.2015.04.019] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/01/2015] [Accepted: 04/08/2015] [Indexed: 01/19/2023]
Abstract
Protein-protein interactions (PPIs) underlie the majority of biological processes, signaling, and disease. Approaches to modulate PPIs with small molecules have therefore attracted increasing interest over the past decade. However, there are a number of challenges inherent in developing small-molecule PPI inhibitors that have prevented these approaches from reaching their full potential. From target validation to small-molecule screening and lead optimization, identifying therapeutically relevant PPIs that can be successfully modulated by small molecules is not a simple task. Following the recent review by Arkin et al., which summarized the lessons learnt from prior successes, we focus in this article on the specific challenges of developing PPI inhibitors and detail the recent advances in chemistry, biology, and computation that facilitate overcoming them. We conclude by providing a perspective on the field and outlining four innovations that we see as key enabling steps for successful development of small-molecule inhibitors targeting PPIs.
Collapse
Affiliation(s)
- Luca Laraia
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK
| | - Grahame McKenzie
- Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK
| | - David R Spring
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Ashok R Venkitaraman
- Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK
| | - David J Huggins
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK; Theory of Condensed Matter Group, Cavendish Laboratory, University of Cambridge, 19 JJ Thomson Avenue, Cambridge CB3 0HE, UK.
| |
Collapse
|
19
|
Magee TV. Progress in discovery of small-molecule modulators of protein-protein interactions via fragment screening. Bioorg Med Chem Lett 2015; 25:2461-8. [PMID: 25971770 DOI: 10.1016/j.bmcl.2015.04.089] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 04/21/2015] [Accepted: 04/27/2015] [Indexed: 11/16/2022]
Abstract
Protein-protein interactions (PPIs) present a formidable challenge to medicinal chemistry. The extended and open nature of many binding sites at protein interfaces has made it difficult to find useful chemical matter by traditional screening methods using standard screening libraries. This Digest focuses on the progress that has been made in discovering small-molecule modulators for a diverse selection of PPI targets using fragment screening and highlights the utility of this strategy in this context.
Collapse
Affiliation(s)
- Thomas V Magee
- Worldwide Medicinal Chemistry, Pfizer Inc, 610 Main Street, Cambridge, MA 02139, USA.
| |
Collapse
|
20
|
Boucher LE, Bosch J. The apicomplexan glideosome and adhesins - Structures and function. J Struct Biol 2015; 190:93-114. [PMID: 25764948 PMCID: PMC4417069 DOI: 10.1016/j.jsb.2015.02.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 02/20/2015] [Accepted: 02/26/2015] [Indexed: 01/10/2023]
Abstract
The apicomplexan family of pathogens, which includes Plasmodium spp. and Toxoplasma gondii, are primarily obligate intracellular parasites and invade multiple cell types. These parasites express extracellular membrane protein receptors, adhesins, to form specific pathogen-host cell interaction complexes. Various adhesins are used to invade a variety of cell types. The receptors are linked to an actomyosin motor, which is part of a complex comprised of many proteins known as the invasion machinery or glideosome. To date, reviews on invasion have focused primarily on the molecular pathways and signals of invasion, with little or no structural information presented. Over 75 structures of parasite receptors and glideosome proteins have been deposited with the Protein Data Bank. These structures include adhesins, motor proteins, bridging proteins, inner membrane complex and cytoskeletal proteins, as well as co-crystal structures with peptides and antibodies. These structures provide information regarding key interactions necessary for target receptor engagement, machinery complex formation, how force is transmitted, and the basis of inhibitory antibodies. Additionally, these structures can provide starting points for the development of antibodies and inhibitory molecules targeting protein-protein interactions, with the aim to inhibit invasion. This review provides an overview of the parasite adhesin protein families, the glideosome components, glideosome architecture, and discuss recent work regarding alternative models.
Collapse
Affiliation(s)
- Lauren E Boucher
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA; Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA.
| | - Jürgen Bosch
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA; Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA.
| |
Collapse
|