1
|
Matlhodi T, Makatsela LP, Dongola TH, Simelane MBC, Shonhai A, Gumede NJ, Mokoena F. Auto QSAR-based active learning docking for hit identification of potential inhibitors of Plasmodium falciparum Hsp90 as antimalarial agents. PLoS One 2024; 19:e0308969. [PMID: 39585817 PMCID: PMC11588265 DOI: 10.1371/journal.pone.0308969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 08/02/2024] [Indexed: 11/27/2024] Open
Abstract
Malaria which is mainly caused by Plasmodium falciparum parasite remains a devastating public health concern, necessitating the need to develop new antimalarial agents. P. falciparum heat shock protein 90 (Hsp90), is indispensable for parasite survival and a promising drug target. Inhibitors targeting the ATP-binding pocket of the N-terminal domain have anti-Plasmodium effects. We proposed a de novo active learning (AL) driven method in tandem with docking to predict inhibitors with unique scaffolds and preferential selectivity towards PfHsp90. Reference compounds, predicted to bind PfHsp90 at the ATP-binding pocket and possessing anti-Plasmodium activities, were used to generate 10,000 unique derivatives and to build the Auto-quantitative structures activity relationships (QSAR) models. Glide docking was performed to predict the docking scores of the derivatives and > 15,000 compounds obtained from the ChEMBL database. Re-iterative training and testing of the models was performed until the optimum Kennel-based Partial Least Square (KPLS) regression model with a regression coefficient R2 = 0.75 for the training set and squared correlation prediction Q2 = 0.62 for the test set reached convergence. Rescoring using induced fit docking and molecular dynamics simulations enabled us to prioritize 15 ATP/ADP-like design ideas for purchase. The compounds exerted moderate activity towards P. falciparum NF54 strain with IC50 values of ≤ 6μM and displayed moderate to weak affinity towards PfHsp90 (KD range: 13.5-19.9μM) comparable to the reported affinity of ADP. The most potent compound was FTN-T5 (PfN54 IC50:1.44μM; HepG2/CHO cells SI≥ 29) which bound to PfHsp90 with moderate affinity (KD:7.7μM), providing a starting point for optimization efforts. Our work demonstrates the great utility of AL for the rapid identification of novel molecules for drug discovery (i.e., hit identification). The potency of FTN-T5 will be critical for designing species-selective inhibitors towards developing more efficient agents against malaria.
Collapse
Affiliation(s)
- Thato Matlhodi
- Department of Biochemistry, Faculty of Natural and Agricultural Science, North-West University, Mmabatho, South Africa
| | - Lisema Patrick Makatsela
- Department of Biochemistry, Faculty of Natural and Agricultural Science, North-West University, Mmabatho, South Africa
| | | | | | - Addmore Shonhai
- Department of Biochemistry & Microbiology, University of Venda, Thohoyandou, South Africa
| | - Njabulo Joyfull Gumede
- Department of Chemical and Physical Sciences, Faculty of Natural Sciences, Walter Sisulu University (WSU), Umthatha, Eastern Cape, South Africa
| | - Fortunate Mokoena
- Department of Biochemistry, Faculty of Natural and Agricultural Science, North-West University, Mmabatho, South Africa
| |
Collapse
|
2
|
Patra J, Arora S, Debnath U, Mahindroo N. In silico studies for improving target selectivity of anti-malarial dual falcipain inhibitors vis-à-vis human cathepsins. J Biomol Struct Dyn 2024:1-20. [PMID: 39552300 DOI: 10.1080/07391102.2024.2427372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 06/21/2024] [Indexed: 11/19/2024]
Abstract
Dual falcipain-2 (FP-2) and falcipain-3 (FP-3) inhibitors, NM12 and NM15, displayed micromolar inhibitions but they exhibit similar binding affinities for the human cathepsins, thus indicating potential toxicity. The current study aims to develop a model to enhance the selectivity of the falcipain inhibitors vis-à-vis human cathepsins using previously identified dual falcipain 2 and 3 inhibitors, NM12 and NM15. To improve the selectivity of NM12 and NM15, analogs with weaker interactions with the conserved residues in the FPs and hCatK were designed while enhancing the unique interactions for the FPs. In silico analysis was carried out in the S2 subsite of both plasmodium and human proteases which is considered the preferred selective site due to the presence of less conserved residues. The Fasta sequence alignment and active/conserved binding site superimposition show that FPs contain acidic polar residues (Asp234 for FP2 and Glu243 for FP3) while hCatK has a neutral hydrophobic residue (Leu209) at the S2 subsite. Therefore, analogs of NM12 and NM15 were designed to enhance affinity and selectivity by improving interactions with these acidic residues while avoiding interactions with hydrophobic residues in hCatK. Newly designed analogs (NM12H and NM15G) show better selectivity as well as binding affinity towards FPs (ΔG of NM12H: -74.49 kcal/mol for FP2, -70.97 kcal/mol for FP3; ΔG of NM15G: -70.09 kcal/mol for FP2, -74.52 kcal/mol for FP3) as compared to NM12 and NM15. Thus, the selectivity and binding affinity against dual falcipains vis-à-vis human cathepsin were improved using molecular dynamic simulations.
Collapse
Affiliation(s)
- Jeevan Patra
- School of Health Sciences and Technology, UPES, Energy Acres, Bidholi, India
| | - Smriti Arora
- School of Health Sciences and Technology, UPES, Energy Acres, Bidholi, India
| | - Utsab Debnath
- School of Health Sciences and Technology, UPES, Energy Acres, Bidholi, India
| | - Neeraj Mahindroo
- School of Health Sciences and Technology, UPES, Energy Acres, Bidholi, India
- School of Health Sciences and Technology, Vishwanath Karad MIT World Peace University, Kothrud, Pune, India
| |
Collapse
|
3
|
Qiao H, Wang Z, Yang H, Xia M, Yang G, Bai F, Wang J, Fang P. Specific glycine-dependent enzyme motion determines the potency of conformation selective inhibitors of threonyl-tRNA synthetase. Commun Biol 2024; 7:867. [PMID: 39014102 PMCID: PMC11252418 DOI: 10.1038/s42003-024-06559-x] [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: 02/26/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024] Open
Abstract
The function of proteins depends on their correct structure and proper dynamics. Understanding the dynamics of target proteins facilitates drug design and development. However, dynamic information is often hidden in the spatial structure of proteins. It is important but difficult to identify the specific residues that play a decisive role in protein dynamics. Here, we report that a critical glycine residue (Gly463) dominates the motion of threonyl-tRNA synthetase (ThrRS) and the sensitivity of the enzyme to antibiotics. Obafluorin (OB), a natural antibiotic, is a novel covalent inhibitor of ThrRS. The binding of OB induces a large conformational change in ThrRS. Through five crystal structures, biochemical and biophysical analyses, and computational simulations, we found that Gly463 plays an important role in the dynamics of ThrRS. Mutating this flexible residue into more rigid residues did not damage the enzyme's three-dimensional structure but significantly improved the thermal stability of the enzyme and suppressed its ability to change conformation. These mutations cause resistance of ThrRS to antibiotics that are conformationally selective, such as OB and borrelidin. This work not only elucidates the molecular mechanism of the self-resistance of OB-producing Pseudomonas fluorescens but also emphasizes the importance of backbone kinetics for aminoacyl-tRNA synthetase-targeting drug development.
Collapse
Affiliation(s)
- Hang Qiao
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, 200032, Shanghai, China
| | - Zilu Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, 200032, Shanghai, China
| | - Hao Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, 201210, Shanghai, China
| | - Mingyu Xia
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, 200032, Shanghai, China
| | - Guang Yang
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-lane Xiangshan, 310024, Hangzhou, China
| | - Fang Bai
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, 201210, Shanghai, China.
- School of Information Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, 201210, Shanghai, China.
- Shanghai Clinical Research and Trial Center, 201210, Shanghai, China.
| | - Jing Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, 200032, Shanghai, China.
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-lane Xiangshan, 310024, Hangzhou, China.
| | - Pengfei Fang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, 200032, Shanghai, China.
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-lane Xiangshan, 310024, Hangzhou, China.
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, 510006, Guangzhou, China.
| |
Collapse
|
4
|
Mansfield CR, Quan B, Chirgwin ME, Eduful B, Hughes PF, Neveu G, Sylvester K, Ryan DH, Kafsack BFC, Haystead TAJ, Leahy JW, Fitzgerald MC, Derbyshire ER. Selective targeting of Plasmodium falciparum Hsp90 disrupts the 26S proteasome. Cell Chem Biol 2024; 31:729-742.e13. [PMID: 38492573 PMCID: PMC11031320 DOI: 10.1016/j.chembiol.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 11/09/2023] [Accepted: 02/22/2024] [Indexed: 03/18/2024]
Abstract
The molecular chaperone heat shock protein 90 (Hsp90) has an essential but largely undefined role in maintaining proteostasis in Plasmodium falciparum, the most lethal malaria parasite. Herein, we identify BX-2819 and XL888 as potent P. falciparum (Pf)Hsp90 inhibitors. Derivatization of XL888's scaffold led to the development of Tropane 1, as a PfHsp90-selective binder with nanomolar affinity. Hsp90 inhibitors exhibit anti-Plasmodium activity against the liver, asexual blood, and early gametocyte life stages. Thermal proteome profiling was implemented to assess PfHsp90-dependent proteome stability, and the proteasome-the main site of cellular protein recycling-was enriched among proteins with perturbed stability upon PfHsp90 inhibition. Subsequent biochemical and cellular studies suggest that PfHsp90 directly promotes proteasome hydrolysis by chaperoning the active 26S complex. These findings expand our knowledge of the PfHsp90-dependent proteome and protein quality control mechanisms in these pathogenic parasites, as well as further characterize this chaperone as a potential antimalarial drug target.
Collapse
Affiliation(s)
- Christopher R Mansfield
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Baiyi Quan
- Department of Chemistry, Duke University, Durham, NC, USA
| | | | - Benjamin Eduful
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Philip F Hughes
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Gaëlle Neveu
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Kayla Sylvester
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Daniel H Ryan
- Department of Chemistry, Duke University, Durham, NC, USA
| | - Björn F C Kafsack
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Timothy A J Haystead
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - James W Leahy
- Department of Chemistry, University of South Florida, Tampa, FL, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Center for Drug Discovery and Innovation, University of South Florida, Tampa, FL, USA
| | | | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA; Department of Chemistry, Duke University, Durham, NC, USA.
| |
Collapse
|
5
|
Mafethe O, Ntseane T, Dongola TH, Shonhai A, Gumede NJ, Mokoena F. Pharmacophore Model-Based Virtual Screening Workflow for Discovery of Inhibitors Targeting Plasmodium falciparum Hsp90. ACS OMEGA 2023; 8:38220-38232. [PMID: 37867657 PMCID: PMC10586269 DOI: 10.1021/acsomega.3c04494] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/07/2023] [Indexed: 10/24/2023]
Abstract
Plasmodium falciparum causes the most lethal and widespread form of malaria. Eradication of malaria remains a priority due to the increasing number of cases of drug resistance. The heat shock protein 90 of P. falciparum (PfHsp90) is a validated drug target essential for parasite survival. Most PfHsp90 inhibitors bind at the ATP binding pocket found in its N-terminal domain, abolishing the chaperone's activities, which leads to parasite death. The challenge is that the NTD of PfHsp90 is highly conserved, and its disruption requires selective inhibitors that can act without causing off-target human Hsp90 activities. We endeavored to discover selective inhibitors of PfHsp90 using pharmacophore modeling, virtual screening protocols, induced fit docking (IFD), and cell-based and biochemical assays. The pharmacophore model (DHHRR), composed of one hydrogen bond donor, two hydrophobic groups, and two aromatic rings, was used to mine commercial databases for initial hits, which were rescored to 20 potential hits using IFD. Eight of these compounds displayed moderate to high activity toward P. falciparum NF54 (i.e., IC50s ranging from 6.0 to 0.14 μM) and averaged >10 in terms of selectivity indices toward CHO and HepG2 cells. Additionally, four compounds inhibited PfHsp90 with greater selectivity than a known inhibitor, harmine, and bound to PfHsp90 with weak to moderate affinity. Our findings support the use of a pharmacophore model to discover diverse chemical scaffolds such as FM2, FM6, F10, and F11 exhibiting anti-Plasmodium activities and serving as valuable new PfHsp90 inhibitors. Optimization of these hits may enable their development into potent leads for future antimalarial drugs.
Collapse
Affiliation(s)
- Ofentse Mafethe
- Department
of Biochemistry, North-West University, Mmabatho 2735, South Africa
| | - Tlhalefo Ntseane
- Department
of Biochemistry, North-West University, Mmabatho 2735, South Africa
| | | | - Addmore Shonhai
- Department
of Biochemistry and Microbiology, University
of Venda, Thohoyandou 0950, South Africa
| | - Njabulo Joyfull Gumede
- Department
of Chemical and Physical Sciences, Faculty of Natural Sciences, Walter Sisulu University (WSU), Private Bag X01, Umthatha, Eastern Cape 4099, South Africa
| | - Fortunate Mokoena
- Department
of Biochemistry, North-West University, Mmabatho 2735, South Africa
| |
Collapse
|
6
|
Edkins AL, Blatch GL. Complementation Assays for Co-chaperone Function. Methods Mol Biol 2023; 2693:105-111. [PMID: 37540430 DOI: 10.1007/978-1-0716-3342-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
The development of mutant microorganisms lacking J domain proteins (JDPs; formerly called Hsp40s) has enabled the development of complementation assays for testing the co-chaperone function of JDPs. In these assays, an exogenously expressed novel JDP is tested for its ability to functionally substitute for a non-expressed or nonfunctional endogenous JDP(s) by reversing a stress phenotype. For example, the in vivo functionality of prokaryotic JDPs can be tested on the basis of their ability to reverse the thermosensitivity of a dnaJ cbpA mutant strain of the bacterium Escherichia coli (OD259). Similarly, the in vivo functionality of eukaryotic JDPs can be assessed in a thermosensitive ydj1 mutant strain of the yeast Saccharomyces cerevisiae (JJ160). Here we outline the use of these thermosensitive microorganisms in complementation assays to functionally characterize a JDP from the bacterium, Agrobacterium tumefaciens (AgtDnaJ), and a JDP from the trypanosomal parasite, Trypanosoma cruzi (TcJ2).
Collapse
Affiliation(s)
- Adrienne L Edkins
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Makhanda, South Africa.
| | - Gregory L Blatch
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Makhanda, South Africa.
- Biomedical Research and Drug Discovery Research Group, Faculty of Health Sciences, Higher Colleges of Technology, Sharjah, United Arab Emirates.
- The University of Notre Dame Australia, Fremantle, WA, Australia.
| |
Collapse
|
7
|
Tassone G, Mazzorana M, Mangani S, Petricci E, Cini E, Giannini G, Pozzi C, Maramai S. Structural Characterization of Human Heat Shock Protein 90 N-Terminal Domain and Its Variants K112R and K112A in Complex with a Potent 1,2,3-Triazole-Based Inhibitor. Int J Mol Sci 2022; 23:ijms23169458. [PMID: 36012721 PMCID: PMC9409116 DOI: 10.3390/ijms23169458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/08/2022] [Accepted: 08/14/2022] [Indexed: 11/18/2022] Open
Abstract
Heat shock protein 90 (Hsp90) is a ubiquitous molecular chaperone that stabilizes client proteins in a folded and functional state. It is composed of two identical and symmetrical subunits and each monomer consists of three domains, the N-terminal (NTD), the middle (MD), and the C-terminal domain (CTD). Since the chaperone activity requires ATP hydrolysis, molecules able to occupy the ATP-binding pocket in the NTD act as Hsp90 inhibitors, leading to client protein degradation and cell death. Therefore, human Hsp90 represents a validated target for developing new anticancer drugs. Since protozoan parasites use their Hsp90 to trigger important transitions between different stages of their life cycle, this protein also represents a profitable target in anti-parasite drug discovery. Nevertheless, the development of molecules able to selectively target the ATP-binding site of protozoan Hsp90 is challenging due to the high homology with the human Hsp90 NTD (hHsp90-NTD). In a previous work, a series of potent Hsp90 inhibitors based on a 1,4,5-trisubstituted 1,2,3-triazole scaffold was developed. The most promising inhibitor of the series, JMC31, showed potent Hsp90 binding and antiproliferative activity in NCI-H460 cells in the low-nanomolar range. In this work, we present the structural characterization of hHsp90-NTD in complex with JMC31 through X-ray crystallography. In addition, to elucidate the role of residue 112 on the ligand binding and its exploitability for the development of selective inhibitors, we investigated the crystal structures of hHsp90-NTD variants (K112R and K112A) in complex with JMC31.
Collapse
Affiliation(s)
- Giusy Tassone
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018–2022, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy
| | - Marco Mazzorana
- Diamond Light Source Ltd., Diamond House, Harwell Science & Innovation Campus, Didcot, Oxfordshire OX11 0DE, UK
- Correspondence: (M.M.); (C.P.); Tel.: +44-01235-778643 (M.M.); +39-0577-232132 (C.P.)
| | - Stefano Mangani
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018–2022, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy
| | - Elena Petricci
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018–2022, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy
| | - Elena Cini
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018–2022, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy
| | | | - Cecilia Pozzi
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018–2022, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy
- Correspondence: (M.M.); (C.P.); Tel.: +44-01235-778643 (M.M.); +39-0577-232132 (C.P.)
| | - Samuele Maramai
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018–2022, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy
| |
Collapse
|
8
|
Hsp90 and Associated Co-Chaperones of the Malaria Parasite. Biomolecules 2022; 12:biom12081018. [PMID: 35892329 PMCID: PMC9332011 DOI: 10.3390/biom12081018] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/16/2022] [Accepted: 07/17/2022] [Indexed: 12/14/2022] Open
Abstract
Heat shock protein 90 (Hsp90) is one of the major guardians of cellular protein homeostasis, through its specialized molecular chaperone properties. While Hsp90 has been extensively studied in many prokaryotic and higher eukaryotic model organisms, its structural, functional, and biological properties in parasitic protozoans are less well defined. Hsp90 collaborates with a wide range of co-chaperones that fine-tune its protein folding pathway. Co-chaperones play many roles in the regulation of Hsp90, including selective targeting of client proteins, and the modulation of its ATPase activity, conformational changes, and post-translational modifications. Plasmodium falciparum is responsible for the most lethal form of human malaria. The survival of the malaria parasite inside the host and the vector depends on the action of molecular chaperones. The major cytosolic P. falciparum Hsp90 (PfHsp90) is known to play an essential role in the development of the parasite, particularly during the intra-erythrocytic stage in the human host. Although PfHsp90 shares significant sequence and structural similarity with human Hsp90, it has several major structural and functional differences. Furthermore, its co-chaperone network appears to be substantially different to that of the human host, with the potential absence of a key homolog. Indeed, PfHsp90 and its interface with co-chaperones represent potential drug targets for antimalarial drug discovery. In this review, we critically summarize the current understanding of the properties of Hsp90, and the associated co-chaperones of the malaria parasite.
Collapse
|
9
|
Stofberg ML, Caillet C, de Villiers M, Zininga T. Inhibitors of the Plasmodium falciparum Hsp90 towards Selective Antimalarial Drug Design: The Past, Present and Future. Cells 2021; 10:2849. [PMID: 34831072 PMCID: PMC8616389 DOI: 10.3390/cells10112849] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
Malaria is still one of the major killer parasitic diseases in tropical settings, posing a public health threat. The development of antimalarial drug resistance is reversing the gains made in attempts to control the disease. The parasite leads a complex life cycle that has adapted to outwit almost all known antimalarial drugs to date, including the first line of treatment, artesunate. There is a high unmet need to develop new strategies and identify novel therapeutics to reverse antimalarial drug resistance development. Among the strategies, here we focus and discuss the merits of the development of antimalarials targeting the Heat shock protein 90 (Hsp90) due to the central role it plays in protein quality control.
Collapse
Affiliation(s)
| | | | | | - Tawanda Zininga
- Department of Biochemistry, Stellenbosch University, Stellenbosch 7600, South Africa; (M.L.S.); (C.C.); (M.d.V.)
| |
Collapse
|
10
|
Mahindra A, Janha O, Mapesa K, Sanchez-Azqueta A, Alam MM, Amambua-Ngwa A, Nwakanma DC, Tobin AB, Jamieson AG. Development of Potent PfCLK3 Inhibitors Based on TCMDC-135051 as a New Class of Antimalarials. J Med Chem 2020; 63:9300-9315. [PMID: 32787140 PMCID: PMC7497403 DOI: 10.1021/acs.jmedchem.0c00451] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Indexed: 12/20/2022]
Abstract
The protein kinase PfCLK3 plays a critical role in the regulation of malarial parasite RNA splicing and is essential for the survival of blood stage Plasmodium falciparum. We recently validated PfCLK3 as a drug target in malaria that offers prophylactic, transmission blocking, and curative potential. Herein, we describe the synthesis of our initial hit TCMDC-135051 (1) and efforts to establish a structure-activity relationship with a 7-azaindole-based series. A total of 14 analogues were assessed in a time-resolved fluorescence energy transfer assay against the full-length recombinant protein kinase PfCLK3, and 11 analogues were further assessed in asexual 3D7 (chloroquine-sensitive) strains of P. falciparum parasites. SAR relating to rings A and B was established. These data together with analysis of activity against parasites collected from patients in the field suggest that TCMDC-135051 (1) is a promising lead compound for the development of new antimalarials with a novel mechanism of action targeting PfCLK3.
Collapse
Affiliation(s)
- Amit Mahindra
- School
of Chemistry, University of Glasgow, Joseph Black Building, University
Avenue, Glasgow G12 8QQ, U.K.
| | - Omar Janha
- Centre
for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, University of Glasgow, Davidson Building, Glasgow G12 8QQ, U.K.
| | - Kopano Mapesa
- School
of Chemistry, University of Glasgow, Joseph Black Building, University
Avenue, Glasgow G12 8QQ, U.K.
| | - Ana Sanchez-Azqueta
- Centre
for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, University of Glasgow, Davidson Building, Glasgow G12 8QQ, U.K.
| | - Mahmood M. Alam
- Wellcome
Centre for Integrative Parasitology and Centre for Translational Pharmacology,
Institute of Infection Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, U.K.
| | - Alfred Amambua-Ngwa
- MRC
Unit The Gambia at LSHTM, Atlantic Boulevard,
Fajara, P. O. Box 273, Banjul, The Gambia
| | - Davis C. Nwakanma
- MRC
Unit The Gambia at LSHTM, Atlantic Boulevard,
Fajara, P. O. Box 273, Banjul, The Gambia
| | - Andrew B. Tobin
- Centre
for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, University of Glasgow, Davidson Building, Glasgow G12 8QQ, U.K.
| | - Andrew G. Jamieson
- School
of Chemistry, University of Glasgow, Joseph Black Building, University
Avenue, Glasgow G12 8QQ, U.K.
| |
Collapse
|
11
|
Makumire S, Zininga T, Vahokoski J, Kursula I, Shonhai A. Biophysical analysis of Plasmodium falciparum Hsp70-Hsp90 organising protein (PfHop) reveals a monomer that is characterised by folded segments connected by flexible linkers. PLoS One 2020; 15:e0226657. [PMID: 32343703 PMCID: PMC7188212 DOI: 10.1371/journal.pone.0226657] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/03/2020] [Indexed: 12/20/2022] Open
Abstract
Plasmodium falciparum causes the most lethal form of malaria. The cooperation of heat shock protein (Hsp) 70 and 90 is thought to facilitate folding of select group of cellular proteins that are crucial for cyto-protection and development of the parasites. Hsp70 and Hsp90 are brought into a functional complex that allows substrate exchange by stress inducible protein 1 (STI1), also known as Hsp70-Hsp90 organising protein (Hop). P. falciparum Hop (PfHop) co-localises and occurs in complex with the parasite cytosolic chaperones, PfHsp70-1 and PfHsp90. Here, we characterised the structure of recombinant PfHop using synchrotron radiation circular dichroism (SRCD) and small-angle X-ray scattering. Structurally, PfHop is a monomeric, elongated but folded protein, in agreement with its predicted TPR domain structure. Using SRCD, we established that PfHop is unstable at temperatures higher than 40°C. This suggests that PfHop is less stable at elevated temperatures compared to its functional partner, PfHsp70-1, that is reportedly stable at temperatures as high as 80°C. These findings contribute towards our understanding of the role of the Hop-mediated functional partnership between Hsp70 and Hsp90.
Collapse
Affiliation(s)
- Stanley Makumire
- Department of Biochemistry, School of Mathematical & Natural Sciences, University of Venda, Thohoyandou, South Africa
| | - Tawanda Zininga
- Department of Biochemistry, School of Mathematical & Natural Sciences, University of Venda, Thohoyandou, South Africa
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Juha Vahokoski
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Inari Kursula
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Biocenter Oulu & Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Addmore Shonhai
- Department of Biochemistry, School of Mathematical & Natural Sciences, University of Venda, Thohoyandou, South Africa
- * E-mail:
| |
Collapse
|
12
|
Taldone T, Wang T, Rodina A, Pillarsetty NVK, Digwal CS, Sharma S, Yan P, Joshi S, Pagare PP, Bolaender A, Roboz GJ, Guzman ML, Chiosis G. A Chemical Biology Approach to the Chaperome in Cancer-HSP90 and Beyond. Cold Spring Harb Perspect Biol 2020; 12:a034116. [PMID: 30936118 PMCID: PMC6773535 DOI: 10.1101/cshperspect.a034116] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer is often associated with alterations in the chaperome, a collection of chaperones, cochaperones, and other cofactors. Changes in the expression levels of components of the chaperome, in the interaction strength among chaperome components, alterations in chaperome constituency, and in the cellular location of chaperome members, are all hallmarks of cancer. Here we aim to provide an overview on how chemical biology has played a role in deciphering such complexity in the biology of the chaperome in cancer and in other diseases. The focus here is narrow and on pathologic changes in the chaperome executed by enhancing the interaction strength between components of distinct chaperome pathways, specifically between those of HSP90 and HSP70 pathways. We will review chemical tools and chemical probe-based assays, with a focus on HSP90. We will discuss how kinetic binding, not classical equilibrium binding, is most appropriate in the development of drugs and probes for the chaperome in disease. We will then present our view on how chaperome inhibitors may become potential drugs and diagnostics in cancer.
Collapse
Affiliation(s)
- Tony Taldone
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Tai Wang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Anna Rodina
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | | | - Chander S Digwal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Pengrong Yan
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Suhasini Joshi
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Piyusha P Pagare
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Alexander Bolaender
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Gail J Roboz
- Division of Hematology and Medical Oncology, Leukemia Program, Weill Cornell Medicine/New York-Presbyterian Hospital, New York, New York 10065
| | - Monica L Guzman
- Division of Hematology and Medical Oncology, Leukemia Program, Weill Cornell Medicine/New York-Presbyterian Hospital, New York, New York 10065
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
13
|
Zininga T, Shonhai A. Small Molecule Inhibitors Targeting the Heat Shock Protein System of Human Obligate Protozoan Parasites. Int J Mol Sci 2019; 20:E5930. [PMID: 31775392 PMCID: PMC6929125 DOI: 10.3390/ijms20235930] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/29/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022] Open
Abstract
Obligate protozoan parasites of the kinetoplastids and apicomplexa infect human cells to complete their life cycles. Some of the members of these groups of parasites develop in at least two systems, the human host and the insect vector. Survival under the varied physiological conditions associated with the human host and in the arthropod vectors requires the parasites to modulate their metabolic complement in order to meet the prevailing conditions. One of the key features of these parasites essential for their survival and host infectivity is timely expression of various proteins. Even more importantly is the need to keep their proteome functional by maintaining its functional capabilities in the wake of physiological changes and host immune responses. For this reason, molecular chaperones (also called heat shock proteins)-whose role is to facilitate proteostasis-play an important role in the survival of these parasites. Heat shock protein 90 (Hsp90) and Hsp70 are prominent molecular chaperones that are generally induced in response to physiological stress. Both Hsp90 and Hsp70 members are functionally regulated by nucleotides. In addition, Hsp70 and Hsp90 cooperate to facilitate folding of some key proteins implicated in cellular development. In addition, Hsp90 and Hsp70 individually interact with other accessory proteins (co-chaperones) that regulate their functions. The dependency of these proteins on nucleotide for their chaperone function presents an Achille's heel, as inhibitors that mimic ATP are amongst potential therapeutic agents targeting their function in obligate intracellular human parasites. Most of the promising small molecule inhibitors of parasitic heat shock proteins are either antibiotics or anticancer agents, whose repurposing against parasitic infections holds prospects. Both cancer cells and obligate human parasites depend upon a robust protein quality control system to ensure their survival, and hence, both employ a competent heat shock machinery to this end. Furthermore, some inhibitors that target chaperone and co-chaperone networks also offer promising prospects as antiparasitic agents. The current review highlights the progress made so far in design and application of small molecule inhibitors against obligate intracellular human parasites of the kinetoplastida and apicomplexan kingdoms.
Collapse
Affiliation(s)
| | - Addmore Shonhai
- Department of Biochemistry, School of Mathematical and Natural Sciences, University of Venda, Thohoyandou 0950, South Africa;
| |
Collapse
|
14
|
Anas M, Kumari V, Gupta N, Dube A, Kumar N. Protein quality control machinery in intracellular protozoan parasites: hopes and challenges for therapeutic targeting. Cell Stress Chaperones 2019; 24:891-904. [PMID: 31228085 PMCID: PMC6717229 DOI: 10.1007/s12192-019-01016-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/08/2019] [Accepted: 06/10/2019] [Indexed: 01/28/2023] Open
Abstract
Intracellular protozoan parasites have evolved an efficient protein quality control (PQC) network comprising protein folding and degradation machineries that protect the parasite's proteome from environmental perturbations and threats posed by host immune surveillance. Interestingly, the components of PQC machinery in parasites have acquired sequence insertions which may provide additional interaction interfaces and diversify the repertoire of their biological roles. However, the auxiliary functions of PQC machinery remain poorly explored in parasite. A comprehensive understanding of this critical machinery may help to identify robust biological targets for new drugs against acute or latent and drug-resistant infections. Here, we review the dynamic roles of PQC machinery in creating a safe haven for parasite survival in hostile environments, serving as a metabolic sensor to trigger transformation into phenotypically distinct stages, acting as a lynchpin for trafficking of parasite cargo across host membrane for immune evasion and serving as an evolutionary capacitor to buffer mutations and drug-induced proteotoxicity. Versatile roles of PQC machinery open avenues for exploration of new drug targets for anti-parasitic intervention and design of strategies for identification of potential biomarkers for point-of-care diagnosis.
Collapse
Affiliation(s)
- Mohammad Anas
- Department of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Varsha Kumari
- Department of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Niharika Gupta
- Department of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Anuradha Dube
- Department of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Niti Kumar
- Academy of Scientific and Innovative Research (AcSIR), Delhi, India.
| |
Collapse
|
15
|
Hollingsworth SA, Kelly B, Valant C, Michaelis JA, Mastromihalis O, Thompson G, Venkatakrishnan AJ, Hertig S, Scammells PJ, Sexton PM, Felder CC, Christopoulos A, Dror RO. Cryptic pocket formation underlies allosteric modulator selectivity at muscarinic GPCRs. Nat Commun 2019; 10:3289. [PMID: 31337749 PMCID: PMC6650467 DOI: 10.1038/s41467-019-11062-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 06/20/2019] [Indexed: 01/27/2023] Open
Abstract
Allosteric modulators are highly desirable as drugs, particularly for G-protein-coupled receptor (GPCR) targets, because allosteric drugs can achieve selectivity between closely related receptors. The mechanisms by which allosteric modulators achieve selectivity remain elusive, however, particularly given recent structures that reveal similar allosteric binding sites across receptors. Here we show that positive allosteric modulators (PAMs) of the M1 muscarinic acetylcholine receptor (mAChR) achieve exquisite selectivity by occupying a dynamic pocket absent in existing crystal structures. This cryptic pocket forms far more frequently in molecular dynamics simulations of the M1 mAChR than in those of other mAChRs. These observations reconcile mutagenesis data that previously appeared contradictory. Further mutagenesis experiments validate our prediction that preventing cryptic pocket opening decreases the affinity of M1-selective PAMs. Our findings suggest opportunities for the design of subtype-specific drugs exploiting cryptic pockets that open in certain receptors but not in other receptors with nearly identical static structures.
Collapse
Affiliation(s)
- Scott A Hollingsworth
- Departments of Computer Science, Molecular and Cellular Physiology, and Structural Biology, and Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, 94305, USA
- Merck & Co., Boston, MA, 02110, USA
| | - Brendan Kelly
- Departments of Computer Science, Molecular and Cellular Physiology, and Structural Biology, and Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Celine Valant
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, 3052, Australia
| | - Jordan Arthur Michaelis
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, 3052, Australia
| | - Olivia Mastromihalis
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, 3052, Australia
| | - Geoff Thompson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, 3052, Australia
| | - A J Venkatakrishnan
- Departments of Computer Science, Molecular and Cellular Physiology, and Structural Biology, and Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Samuel Hertig
- Departments of Computer Science, Molecular and Cellular Physiology, and Structural Biology, and Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Peter J Scammells
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, 3052, Australia
| | - Patrick M Sexton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, 3052, Australia
| | - Christian C Felder
- Eli Lilly and Co., Neuroscience, Lilly Corporate Center, Indianapolis, IN, 46285, USA
- Karuna Pharmaceuticals, Inc., South San Francisco, CA, 94080, USA
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, 3052, Australia.
| | - Ron O Dror
- Departments of Computer Science, Molecular and Cellular Physiology, and Structural Biology, and Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
16
|
Diao PC, Hu MJ, Yang HK, You WW, Zhao PL. Facile one-pot synthesis, antiproliferative evaluation and structure-activity relationships of 3-amino-1H-indoles and 3-amino-1H-7-azaindoles. Bioorg Chem 2019; 88:102914. [DOI: 10.1016/j.bioorg.2019.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/26/2019] [Accepted: 04/06/2019] [Indexed: 12/31/2022]
|
17
|
Engel JA, Norris EL, Gilson P, Przyborski J, Shonhai A, Blatch GL, Skinner-Adams TS, Gorman J, Headlam M, Andrews KT. Proteomic analysis of Plasmodium falciparum histone deacetylase 1 complex proteins. Exp Parasitol 2019; 198:7-16. [PMID: 30682336 DOI: 10.1016/j.exppara.2019.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 12/01/2018] [Accepted: 01/20/2019] [Indexed: 01/12/2023]
Abstract
Plasmodium falciparum histone deacetylases (PfHDACs) are an important class of epigenetic regulators that alter protein lysine acetylation, contributing to regulation of gene expression and normal parasite growth and development. PfHDACs are therefore under investigation as drug targets for malaria. Despite this, our understanding of the biological roles of these enzymes is only just beginning to emerge. In higher eukaryotes, HDACs function as part of multi-protein complexes and act on both histone and non-histone substrates. Here, we present a proteomics analysis of PfHDAC1 immunoprecipitates, identifying 26 putative P. falciparum complex proteins in trophozoite-stage asexual intraerythrocytic parasites. The co-migration of two of these (P. falciparum heat shock proteins 70-1 and 90) with PfHDAC1 was validated using Blue Native PAGE combined with Western blot. These data provide a snapshot of possible PfHDAC1 interactions and a starting point for future studies focused on elucidating the broader function of PfHDACs in Plasmodium parasites.
Collapse
Affiliation(s)
- Jessica A Engel
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Emma L Norris
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Paul Gilson
- Burnet Institute, Monash University, Victoria, Australia
| | - Jude Przyborski
- Centre of Infectious Diseases, Parasitology, University Hospital Heidelberg, Germany
| | - Addmore Shonhai
- Biochemistry Department, University of Venda, Thohoyandou, South Africa
| | - Gregory L Blatch
- The Vice Chancellery, The University of Notre Dame Australia, Fremantle, WA, Australia
| | - Tina S Skinner-Adams
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Jeffrey Gorman
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | | | - Katherine T Andrews
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia.
| |
Collapse
|
18
|
Mbaba M, de la Mare JA, Sterrenberg JN, Kajewole D, Maharaj S, Edkins AL, Isaacs M, Hoppe HC, Khanye SD. Novobiocin-ferrocene conjugates possessing anticancer and antiplasmodial activity independent of HSP90 inhibition. J Biol Inorg Chem 2018; 24:139-149. [PMID: 30542925 DOI: 10.1007/s00775-018-1634-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/04/2018] [Indexed: 12/22/2022]
Abstract
A series of tailored novobiocin-ferrocene conjugates was prepared in moderate yields and investigated for in vitro anticancer and antiplasmodial activity against the MDA-MB-231 breast cancer line and Plasmodium falciparum 3D7 strain, respectively. While the target compounds displayed moderate anticancer activity against the breast cancer cell line with IC50 values in the mid-micromolar range, compounds 10a-c displayed promising antiplasmodial activity as low as 0.889 µM. Furthermore, the most promising compounds were tested for inhibitory effects against a postulated target, heat shock protein 90 (Hsp90). A selection of tailored novobiocin derivatives bearing the organometallic ferrocene unit were synthesized and characterized by common spectroscopic techniques. The target compounds were investigated for in vitro anticancer and antimalarial activity against the MDA-MB-231 breast cancer cell line and Plasmodium falciparum 3D7 strain, respectively.
Collapse
Affiliation(s)
- Mziyanda Mbaba
- Department of Chemistry, Faculty of Science, Rhodes University, Grahamstown, 6140, South Africa.
| | - Jo-Anne de la Mare
- Department of Biochemistry and Microbiology, Faculty of Science, Rhodes University, Grahamstown, 6140, South Africa.,Biomedical Biotechnology Research Unit (BioBRU), Rhodes University, Grahamstown, 6140, South Africa.,Centre for Chemico- and Biomedical Research, Rhodes University, Grahamstown, 6140, South Africa
| | - Jason N Sterrenberg
- Department of Biochemistry and Microbiology, Faculty of Science, Rhodes University, Grahamstown, 6140, South Africa.,Biomedical Biotechnology Research Unit (BioBRU), Rhodes University, Grahamstown, 6140, South Africa
| | - Deborah Kajewole
- Department of Biochemistry and Microbiology, Faculty of Science, Rhodes University, Grahamstown, 6140, South Africa.,Biomedical Biotechnology Research Unit (BioBRU), Rhodes University, Grahamstown, 6140, South Africa
| | - Shantal Maharaj
- Department of Biochemistry and Microbiology, Faculty of Science, Rhodes University, Grahamstown, 6140, South Africa.,Biomedical Biotechnology Research Unit (BioBRU), Rhodes University, Grahamstown, 6140, South Africa
| | - Adrienne L Edkins
- Department of Biochemistry and Microbiology, Faculty of Science, Rhodes University, Grahamstown, 6140, South Africa.,Biomedical Biotechnology Research Unit (BioBRU), Rhodes University, Grahamstown, 6140, South Africa.,Centre for Chemico- and Biomedical Research, Rhodes University, Grahamstown, 6140, South Africa
| | - Michelle Isaacs
- Centre for Chemico- and Biomedical Research, Rhodes University, Grahamstown, 6140, South Africa
| | - Heinrich C Hoppe
- Department of Biochemistry and Microbiology, Faculty of Science, Rhodes University, Grahamstown, 6140, South Africa.,Biomedical Biotechnology Research Unit (BioBRU), Rhodes University, Grahamstown, 6140, South Africa.,Centre for Chemico- and Biomedical Research, Rhodes University, Grahamstown, 6140, South Africa
| | - Setshaba D Khanye
- Department of Chemistry, Faculty of Science, Rhodes University, Grahamstown, 6140, South Africa. .,Centre for Chemico- and Biomedical Research, Rhodes University, Grahamstown, 6140, South Africa. .,Faculty of Pharmacy, Rhodes University, Grahamstown, 6140, South Africa.
| |
Collapse
|
19
|
Wang T, Rodina A, Dunphy MP, Corben A, Modi S, Guzman ML, Gewirth DT, Chiosis G. Chaperome heterogeneity and its implications for cancer study and treatment. J Biol Chem 2018; 294:2162-2179. [PMID: 30409908 DOI: 10.1074/jbc.rev118.002811] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The chaperome is the collection of proteins in the cell that carry out molecular chaperoning functions. Changes in the interaction strength between chaperome proteins lead to an assembly that is functionally and structurally distinct from each constituent member. In this review, we discuss the epichaperome, the cellular network that forms when the chaperome components of distinct chaperome machineries come together as stable, functionally integrated, multimeric complexes. In tumors, maintenance of the epichaperome network is vital for tumor survival, rendering them vulnerable to therapeutic interventions that target critical epichaperome network components. We discuss how the epichaperome empowers an approach for precision medicine cancer trials where a new target, biomarker, and relevant drug candidates can be correlated and integrated. We introduce chemical biology methods to investigate the heterogeneity of the chaperome in a given cellular context. Lastly, we discuss how ligand-protein binding kinetics are more appropriate than equilibrium binding parameters to characterize and unravel chaperome targeting in cancer and to gauge the selectivity of ligands for specific tumor-associated chaperome pools.
Collapse
Affiliation(s)
- Tai Wang
- From the Chemical Biology Program and
| | | | | | - Adriana Corben
- the Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Shanu Modi
- Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Monica L Guzman
- Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, New York 10065, and
| | - Daniel T Gewirth
- the Hauptman-Woodward Medical Research Institute, Buffalo, New York 14203
| | - Gabriela Chiosis
- From the Chemical Biology Program and .,Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
20
|
Tassone G, Mangani S, Botta M, Pozzi C. Probing the role of Arg97 in Heat shock protein 90 N-terminal domain from the parasite Leishmania braziliensis through site-directed mutagenesis on the human counterpart. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1866:1190-1198. [PMID: 30248409 DOI: 10.1016/j.bbapap.2018.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/10/2018] [Accepted: 09/17/2018] [Indexed: 10/28/2022]
Abstract
In Brazil, the mucocutaneous form of leishmaniasis, caused by the parasite Leishmania braziliensis, is a widespread and very challenging disease responsible for disfiguration and, in the most severe cases, death. Heat shock protein 90 (Hsp90) is a ubiquitous molecular chaperone playing a pivotal role in the folding process of client proteins, and therefore its activity is fundamental for cell survival and proliferation. Since the chaperone activity requires ATP hydrolysis, molecules able to occupy the ATP binding pocket in the protein N-terminal domain (NTD) act as Hsp90 inhibitors. The development of selective molecules targeting the ATPase site of protozoan Hsp90 is tricky for the high homology with the human Hsp90 NTD (hNTD). Notably, only the human Lys112 is replaced by Arg97 in the L. braziliensis enzyme. Recently, this difference has been probed to design selective inhibitors targeting parasite Hsp90s. Here, a reliable protocol for expression and purification of LbHsp90-NTD (LbNTD) was developed but its structural characterization was unsuccessful. The role of Arg97 in LbNTD was hence probed by means of the "leishmanized" K112R variant of hNTDα. To deeply investigate the role of this residue, also the hNTDα K112A variant was generated. Structural studies performed on hNTDα and its variants using various ADP and ATP analogues and cAMP revealed that this residue is not crucial for nucleotide binding. This finding strongly suggests that Arg97 in LbNTD and more generally the conserved arginine residue in parasite Hsp90s are not exploitable for the development of selective inhibitors.
Collapse
Affiliation(s)
- Giusy Tassone
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro, 2, 53100 Siena (SI), Italy
| | - Stefano Mangani
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro, 2, 53100 Siena (SI), Italy
| | - Maurizio Botta
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro, 2, 53100 Siena (SI), Italy; Lead Discovery Siena S.r.l., Castelnuovo Berardenga, Siena, Italy; Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Cecilia Pozzi
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro, 2, 53100 Siena (SI), Italy.
| |
Collapse
|
21
|
Quiliano M, Pabón A, Moles E, Bonilla-Ramirez L, Fabing I, Fong KY, Nieto-Aco DA, Wright DW, Pizarro JC, Vettorazzi A, López de Cerain A, Deharo E, Fernández-Busquets X, Garavito G, Aldana I, Galiano S. Structure-activity relationship of new antimalarial 1-aryl-3-susbtituted propanol derivatives: Synthesis, preliminary toxicity profiling, parasite life cycle stage studies, target exploration, and targeted delivery. Eur J Med Chem 2018; 152:489-514. [PMID: 29754074 DOI: 10.1016/j.ejmech.2018.04.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 01/09/2023]
Abstract
Design, synthesis, structure-activity relationship, cytotoxicity studies, in silico drug-likeness, genotoxicity screening, and in vivo studies of new 1-aryl-3-substituted propanol derivatives led to the identification of nine compounds with promising in vitro (55, 56, 61, 64, 66, and 70-73) and in vivo (66 and 72) antimalarial profiles against Plasmodium falciparum and Plasmodium berghei. Compounds 55, 56, 61, 64, 66 and 70-73 exhibited potent antiplasmodial activity against chloroquine-resistant strain FCR-3 (IC50s < 0.28 μM), and compounds 55, 56, 64, 70, 71, and 72 showed potent biological activity in chloroquine-sensitive and multidrug-resistant strains (IC50s < 0.7 μM for 3D7, D6, FCR-3 and C235). All of these compounds share appropriate drug-likeness profiles and adequate selectivity indexes (77 < SI < 184) as well as lack genotoxicity. In vivo efficacy tests in a mouse model showed compounds 66 and 72 to be promising candidates as they exhibited significant parasitemia reductions of 96.4% and 80.4%, respectively. Additional studies such as liver stage and sporogony inhibition, target exploration of heat shock protein 90 of P. falciparum, targeted delivery by immunoliposomes, and enantiomer characterization were performed and strongly reinforce the hypothesis of 1-aryl-3-substituted propanol derivatives as promising antimalarial compounds.
Collapse
Affiliation(s)
- Miguel Quiliano
- Universidad de Navarra, Instituto de Salud Tropical (ISTUN), Campus Universitario, 31008 Pamplona, Spain; Universidad de Navarra, Facultad de Farmacia y Nutrición, Departamento de Química Orgánica y Farmacéutica, Campus Universitario, 31008 Pamplona, Spain
| | - Adriana Pabón
- Grupo Malaria, Universidad de Antioquía, Medellín, Colombia
| | - Ernest Moles
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac 10-12, 08028 Barcelona, Spain; Barcelona Institute for Global Health (ISGlobal), Barcelona Center for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona), Rosselló 149-153, 08036 Barcelona, Spain; Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | | | - Isabelle Fabing
- Laboratoire de Synthese et Physicochimie de Molécules d'Intéret Biologique SPCMIB-UMR5068, CNRS - Université Paul Sabatier, 118, route de Narbonne, 31062, Toulouse Cedex 09, France
| | - Kim Y Fong
- Department of Chemistry, Vanderbilt University, Station B 351822, Nashville, TN 37235, USA
| | - Diego A Nieto-Aco
- Universidad de Navarra, Instituto de Salud Tropical (ISTUN), Campus Universitario, 31008 Pamplona, Spain; Universidad de Navarra, Facultad de Farmacia y Nutrición, Departamento de Química Orgánica y Farmacéutica, Campus Universitario, 31008 Pamplona, Spain
| | - David W Wright
- Department of Chemistry, Vanderbilt University, Station B 351822, Nashville, TN 37235, USA
| | - Juan C Pizarro
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University USA; Vector-Borne Infectious Diseases Research Center, Tulane University USA
| | - Ariane Vettorazzi
- Universidad de Navarra, Facultad de Farmacia y Nutrición, Department of Pharmacology and Toxicology, Campus Universitario, 31008 Pamplona, Spain
| | - Adela López de Cerain
- Universidad de Navarra, Facultad de Farmacia y Nutrición, Department of Pharmacology and Toxicology, Campus Universitario, 31008 Pamplona, Spain
| | - Eric Deharo
- UMR 152 PHARMA-DEV, Université Toulouse, IRD, UPS, 31062, Toulouse, France
| | - Xavier Fernández-Busquets
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac 10-12, 08028 Barcelona, Spain; Barcelona Institute for Global Health (ISGlobal), Barcelona Center for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona), Rosselló 149-153, 08036 Barcelona, Spain; Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Giovanny Garavito
- Universidad Nacional de Colombia, Sede Bogotá, Facultad de Ciencias, Departamento de Farmacia (DFUNC), Grupo de investigación FaMeTra (Farmacología de la Medicina tradicional y popular), Carrera 30 45-03, Bogotá D.C., Colombia
| | - Ignacio Aldana
- Universidad de Navarra, Instituto de Salud Tropical (ISTUN), Campus Universitario, 31008 Pamplona, Spain; Universidad de Navarra, Facultad de Farmacia y Nutrición, Departamento de Química Orgánica y Farmacéutica, Campus Universitario, 31008 Pamplona, Spain
| | - Silvia Galiano
- Universidad de Navarra, Instituto de Salud Tropical (ISTUN), Campus Universitario, 31008 Pamplona, Spain; Universidad de Navarra, Facultad de Farmacia y Nutrición, Departamento de Química Orgánica y Farmacéutica, Campus Universitario, 31008 Pamplona, Spain.
| |
Collapse
|
22
|
Posfai D, Eubanks AL, Keim AI, Lu KY, Wang GZ, Hughes PF, Kato N, Haystead TA, Derbyshire ER. Identification of Hsp90 Inhibitors with Anti-Plasmodium Activity. Antimicrob Agents Chemother 2018; 62:e01799-17. [PMID: 29339390 PMCID: PMC5913967 DOI: 10.1128/aac.01799-17] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 01/05/2018] [Indexed: 12/17/2022] Open
Abstract
Malaria remains a global health burden partly due to Plasmodium parasite resistance to first-line therapeutics. The molecular chaperone heat shock protein 90 (Hsp90) has emerged as an essential protein for blood-stage Plasmodium parasites, but details about its function during malaria's elusive liver stage are unclear. We used target-based screens to identify compounds that bind to Plasmodium falciparum and human Hsp90, which revealed insights into chemotypes with species-selective binding. Using cell-based malaria assays, we demonstrate that all identified Hsp90-binding compounds are liver- and blood-stage Plasmodium inhibitors. Additionally, the Hsp90 inhibitor SNX-0723 in combination with the phosphatidylinositol 3-kinase inhibitor PIK-75 synergistically reduces the liver-stage parasite load. Time course inhibition studies with the Hsp90 inhibitors and expression analysis support a role for Plasmodium Hsp90 in late-liver-stage parasite development. Our results suggest that Plasmodium Hsp90 is essential to liver- and blood-stage parasite infections and highlight an attractive route for development of species-selective PfHsp90 inhibitors that may act synergistically in combination therapies to prevent and treat malaria.
Collapse
Affiliation(s)
- Dora Posfai
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Amber L Eubanks
- Department of Chemistry, Duke University, Durham, North Carolina, USA
| | - Allison I Keim
- Department of Chemistry, Duke University, Durham, North Carolina, USA
| | - Kuan-Yi Lu
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Grace Z Wang
- Department of Chemistry, Duke University, Durham, North Carolina, USA
| | - Philip F Hughes
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | | | - Timothy A Haystead
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Chemistry, Duke University, Durham, North Carolina, USA
| |
Collapse
|
23
|
Murillo-Solano C, Dong C, Sanchez CG, Pizarro JC. Identification and characterization of the antiplasmodial activity of Hsp90 inhibitors. Malar J 2017; 16:292. [PMID: 28724415 PMCID: PMC5518105 DOI: 10.1186/s12936-017-1940-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 07/14/2017] [Indexed: 01/13/2023] Open
Abstract
Background The recent reduction in mortality due to malaria is being threatened by the appearance of Plasmodium falciparum parasites that are resistant to artemisinin in Southeast Asia. To limit the impact of resistant parasites and their spread across the world, there is a need to validate anti-malarial drug targets and identify new leads that will serve as foundations for future drug development programmes targeting malaria. Towards that end, the antiplasmodial potential of several Hsp90 inhibitors was characterized. Because, the Hsp90 chaperone has been suggested as a good drug target against multiple parasitic infections including malaria. Results Chemically diverse sets of Hsp90 inhibitors, evaluated in clinical trials as anti-cancer agents, were tested against the malaria parasite. Most of the compounds showed strong antiplasmodial activity in growth inhibition assays against chloroquine sensitive and resistant strains. There was a good agreement between the compound in vitro anti-parasitic activity and their affinity against the Plasmodium chaperone. The two most potent Hsp90 inhibitors also showed cytocidal activity against two P. falciparum strains. Their antiplasmodial activity affected all parasite forms during the malaria blood cycle. However, the compounds activity against the parasite showed no synergy when combined with anti-malarial drugs, like chloroquine or DHA. Discussion The Hsp90 inhibitors anti-parasitic activity correlates with their affinity to their predicted target the P. falciparum chaperone Hsp90. However, the most effective compounds also showed high affinity for a close homologue, Grp94. This association points to a mode of action for Hsp90 inhibitors that correlate compound efficacy with multi-target engagement. Besides their ability to limit parasite replication, two compounds also significantly impacted P. falciparum viability in vitro. Finally, a structural analysis suggests that the best hit represents a promising scaffold to develop parasite specific leads according. Conclusion The results shown that Hsp90 inhibitors are lethal against the malaria parasite. The correlation between biochemical and in vitro data strongly supports Hsp90 as a drug target against the malaria parasite. Furthermore, at least one Hsp90 inhibitor developed as anticancer therapeutics could serve as starting point to generate P. falciparum-specific lead compounds. Electronic supplementary material The online version of this article (doi:10.1186/s12936-017-1940-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claribel Murillo-Solano
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Chunmin Dong
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Cecilia G Sanchez
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Juan C Pizarro
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA. .,Vector-Borne Infectious Diseases Research Center, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
24
|
Goode KM, Petrov DP, Vickman RE, Crist SA, Pascuzzi PE, Ratliff TL, Davisson VJ, Hazbun TR. Targeting the Hsp90 C-terminal domain to induce allosteric inhibition and selective client downregulation. Biochim Biophys Acta Gen Subj 2017; 1861:1992-2006. [PMID: 28495207 DOI: 10.1016/j.bbagen.2017.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 04/20/2017] [Accepted: 05/05/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Inhibition of Hsp90 is desirable due to potential downregulation of oncogenic clients. Early generation inhibitors bind to the N-terminal domain (NTD) but C-terminal domain (CTD) inhibitors are a promising class because they do not induce a heat shock response. Here we present a new structural class of CTD binding molecules with a unique allosteric inhibition mechanism. METHODS A hit molecule, NSC145366, and structurally similar probes were assessed for inhibition of Hsp90 activities. A ligand-binding model was proposed indicating a novel Hsp90 CTD binding site. Client protein downregulation was also determined. RESULTS NSC145366 interacts with the Hsp90 CTD and has anti-proliferative activity in tumor cell lines (GI50=0.2-1.9μM). NSC145366 increases Hsp90 oligomerization resulting in allosteric inhibition of NTD ATPase activity (IC50=119μM) but does not compete with NTD or CTD-ATP binding. Treatment of LNCaP prostate tumor cells resulted in selective client protein downregulation including AR and BRCA1 but without a heat shock response. Analogs had similar potencies in ATPase and chaperone activity assays and variable effects on oligomerization. In silico modeling predicted a binding site at the CTD dimer interface distinct from the nucleotide-binding site. CONCLUSIONS A set of symmetrical scaffold molecules with bisphenol A cores induced allosteric inhibition of Hsp90. Experimental evidence and molecular modeling suggest that the binding site is independent of the CTD-ATP site and consistent with unique induction of allosteric effects. GENERAL SIGNIFICANCE Allosteric inhibition of Hsp90 via a mechanism used by the NSC145366-based probes is a promising avenue for selective oncogenic client downregulation.
Collapse
Affiliation(s)
- Kourtney M Goode
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Dino P Petrov
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Renee E Vickman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Scott A Crist
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Pete E Pascuzzi
- Purdue University Libraries Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Tim L Ratliff
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - V Jo Davisson
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Tony R Hazbun
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
25
|
Wang T, Mäser P, Picard D. Inhibition of Plasmodium falciparum Hsp90 Contributes to the Antimalarial Activities of Aminoalcohol-carbazoles. J Med Chem 2016; 59:6344-52. [PMID: 27312008 DOI: 10.1021/acs.jmedchem.6b00591] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Malaria caused by the protozoan parasite Plasmodium falciparum (Pf) remains a major public health problem throughout the developing world. One molecular target that should receive more attention is the molecular chaperone Hsp90. It is essential and highly conserved in all eukaryotes, including in protozoan parasites. We have identified an amino-alcohol carbazole (N-CBZ) as a PfHsp90-selective inhibitor by virtually docking a large set of antimalarial compounds, previously found in a phenotypic screen, into a PfHsp90-specific pocket. By correlating the ability of 30 additional N-CBZ derivatives to bind directly to PfHsp90 with their Pf-inhibitory activity, we found that these types of compounds are more likely to inhibit Pf growth if they bind PfHsp90. For plausible targets such as PfHsp90, our workflow may help identifying the molecular target for compounds found by screening large chemical libraries for a desired biological effect and, conversely, ensuring biological effectiveness for compounds affecting a particular target.
Collapse
Affiliation(s)
- Tai Wang
- Département de Biologie Cellulaire, Université de Genève , Sciences III, 30 Quai Ernest-Ansermet, CH-1211 Genève 4, Switzerland
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute , Socinstrasse 57, CH-4051 Basel, Switzerland.,University of Basel , Petersplatz 1, CH-4001 Basel, Switzerland
| | - Didier Picard
- Département de Biologie Cellulaire, Université de Genève , Sciences III, 30 Quai Ernest-Ansermet, CH-1211 Genève 4, Switzerland
| |
Collapse
|
26
|
Abstract
The dynamics of protein binding pockets are crucial for their interaction specificity. Structural flexibility allows proteins to adapt to their individual molecular binding partners and facilitates the binding process. This implies the necessity to consider protein internal motion in determining and predicting binding properties and in designing new binders. Although accounting for protein dynamics presents a challenge for computational approaches, it expands the structural and physicochemical space for compound design and thus offers the prospect of improved binding specificity and selectivity. A cavity on the surface or in the interior of a protein that possesses suitable properties for binding a ligand is usually referred to as a binding pocket. The set of amino acid residues around a binding pocket determines its physicochemical characteristics and, together with its shape and location in a protein, defines its functionality. Residues outside the binding site can also have a long-range effect on the properties of the binding pocket. Cavities with similar functionalities are often conserved across protein families. For example, enzyme active sites are usually concave surfaces that present amino acid residues in a suitable configuration for binding low molecular weight compounds. Macromolecular binding pockets, on the other hand, are located on the protein surface and are often shallower. The mobility of proteins allows the opening, closing, and adaptation of binding pockets to regulate binding processes and specific protein functionalities. For example, channels and tunnels can exist permanently or transiently to transport compounds to and from a binding site. The influence of protein flexibility on binding pockets can vary from small changes to an already existent pocket to the formation of a completely new pocket. Here, we review recent developments in computational methods to detect and define binding pockets and to study pocket dynamics. We introduce five different classes of protein pocket dynamics: (1) appearance/disappearance of a subpocket in an existing pocket; (2) appearance/disappearance of an adjacent pocket on the protein surface in the direct vicinity of an already existing pocket; (3) pocket breathing, which may be caused by side-chain fluctuations or backbone or interdomain vibrational motion; (4) opening/closing of a channel or tunnel, connecting a pocket inside the protein with solvent, including lid motion; and (5) the appearance/disappearance of an allosteric pocket at a site on a protein distinct from an already existing pocket with binding of a ligand to the allosteric binding site affecting the original pocket. We suggest that the class of pocket dynamics, as well as the type and extent of protein motion affecting the binding pocket, should be factors considered in choosing the most appropriate computational approach to study a given binding pocket. Furthermore, we examine the relationship between pocket dynamics classes and induced fit, conformational selection, and gating models of ligand binding on binding kinetics and thermodynamics. We discuss the implications of protein binding pocket dynamics for drug design and conclude with potential future directions for computational analysis of protein binding pocket dynamics.
Collapse
Affiliation(s)
- Antonia Stank
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany
| | - Daria B. Kokh
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany
| | - Jonathan C. Fuller
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany
| | - Rebecca C. Wade
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany
- Center
for Molecular Biology of the University of Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| |
Collapse
|
27
|
Njogu PM, Guantai EM, Pavadai E, Chibale K. Computer-Aided Drug Discovery Approaches against the Tropical Infectious Diseases Malaria, Tuberculosis, Trypanosomiasis, and Leishmaniasis. ACS Infect Dis 2016; 2:8-31. [PMID: 27622945 DOI: 10.1021/acsinfecdis.5b00093] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Despite the tremendous improvement in overall global health heralded by the adoption of the Millennium Declaration in the year 2000, tropical infections remain a major health problem in the developing world. Recent estimates indicate that the major tropical infectious diseases, namely, malaria, tuberculosis, trypanosomiasis, and leishmaniasis, account for more than 2.2 million deaths and a loss of approximately 85 million disability-adjusted life years annually. The crucial role of chemotherapy in curtailing the deleterious health and economic impacts of these infections has invigorated the search for new drugs against tropical infectious diseases. The research efforts have involved increased application of computational technologies in mainstream drug discovery programs at the hit identification, hit-to-lead, and lead optimization stages. This review highlights various computer-aided drug discovery approaches that have been utilized in efforts to identify novel antimalarial, antitubercular, antitrypanosomal, and antileishmanial agents. The focus is largely on developments over the past 5 years (2010-2014).
Collapse
Affiliation(s)
- Peter M. Njogu
- Department of Pharmaceutical Chemistry and ‡Division of Pharmacology, School of Pharmacy, University of Nairobi, P.O. Box 19676-00202, Nairobi, Kenya
- Department of Chemistry, ⊗Institute of Infectious
Disease and Molecular Medicine, and ΘSouth African Medical Research Council Drug
Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
| | - Eric M. Guantai
- Department of Pharmaceutical Chemistry and ‡Division of Pharmacology, School of Pharmacy, University of Nairobi, P.O. Box 19676-00202, Nairobi, Kenya
- Department of Chemistry, ⊗Institute of Infectious
Disease and Molecular Medicine, and ΘSouth African Medical Research Council Drug
Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
| | - Elumalai Pavadai
- Department of Pharmaceutical Chemistry and ‡Division of Pharmacology, School of Pharmacy, University of Nairobi, P.O. Box 19676-00202, Nairobi, Kenya
- Department of Chemistry, ⊗Institute of Infectious
Disease and Molecular Medicine, and ΘSouth African Medical Research Council Drug
Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
| | - Kelly Chibale
- Department of Pharmaceutical Chemistry and ‡Division of Pharmacology, School of Pharmacy, University of Nairobi, P.O. Box 19676-00202, Nairobi, Kenya
- Department of Chemistry, ⊗Institute of Infectious
Disease and Molecular Medicine, and ΘSouth African Medical Research Council Drug
Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
28
|
Aneja B, Kumar B, Jairajpuri MA, Abid M. A structure guided drug-discovery approach towards identification of Plasmodium inhibitors. RSC Adv 2016. [DOI: 10.1039/c5ra19673f] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
This article provides a comprehensive review of inhibitors from natural, semisynthetic or synthetic sources against key targets ofPlasmodium falciparum.
Collapse
Affiliation(s)
- Babita Aneja
- Medicinal Chemistry Lab
- Department of Biosciences
- Jamia Millia Islamia (A Central University)
- New Delhi 110025
- India
| | - Bhumika Kumar
- Medicinal Chemistry Lab
- Department of Biosciences
- Jamia Millia Islamia (A Central University)
- New Delhi 110025
- India
| | - Mohamad Aman Jairajpuri
- Protein Conformation and Enzymology Lab
- Department of Biosciences
- Jamia Millia Islamia (A Central University)
- New Delhi 110025
- India
| | - Mohammad Abid
- Medicinal Chemistry Lab
- Department of Biosciences
- Jamia Millia Islamia (A Central University)
- New Delhi 110025
- India
| |
Collapse
|
29
|
Imidazopyridazine Inhibitors of Plasmodium falciparum Calcium-Dependent Protein Kinase 1 Also Target Cyclic GMP-Dependent Protein Kinase and Heat Shock Protein 90 To Kill the Parasite at Different Stages of Intracellular Development. Antimicrob Agents Chemother 2015; 60:1464-75. [PMID: 26711771 PMCID: PMC4775997 DOI: 10.1128/aac.01748-15] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 12/01/2015] [Indexed: 01/24/2023] Open
Abstract
Imidazopyridazine compounds are potent, ATP-competitive inhibitors of calcium-dependent protein kinase 1 (CDPK1) and of Plasmodium falciparum parasite growth in vitro. Here, we show that these compounds can be divided into two classes depending on the nature of the aromatic linker between the core and the R2 substituent group. Class 1 compounds have a pyrimidine linker and inhibit parasite growth at late schizogony, whereas class 2 compounds have a nonpyrimidine linker and inhibit growth in the trophozoite stage, indicating different modes of action for the two classes. The compounds also inhibited cyclic GMP (cGMP)-dependent protein kinase (PKG), and their potency against this enzyme was greatly reduced by substitution of the enzyme's gatekeeper residue at the ATP binding site. The effectiveness of the class 1 compounds against a parasite line expressing the modified PKG was also substantially reduced, suggesting that these compounds kill the parasite primarily through inhibition of PKG rather than CDPK1. HSP90 was identified as a binding partner of class 2 compounds, and a representative compound bound to the ATP binding site in the N-terminal domain of HSP90. Reducing the size of the gatekeeper residue of CDPK1 enabled inhibition of the enzyme by bumped kinase inhibitors; however, a parasite line expressing the modified enzyme showed no change in sensitivity to these compounds. Taken together, these findings suggest that CDPK1 may not be a suitable target for further inhibitor development and that the primary mechanism through which the imidazopyridazines kill parasites is by inhibition of PKG or HSP90.
Collapse
|
30
|
Faya N, Penkler DL, Tastan Bishop Ö. Human, vector and parasite Hsp90 proteins: A comparative bioinformatics analysis. FEBS Open Bio 2015; 5:916-27. [PMID: 26793431 PMCID: PMC4688443 DOI: 10.1016/j.fob.2015.11.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/23/2015] [Accepted: 11/09/2015] [Indexed: 12/27/2022] Open
Abstract
Cytosolic Hsp90s are more conserved than those from mitochondrial and ER. Cell environment plays a role in the overall physicochemical properties of Hsp90s. Serine and tyrosine are favored phosphorylated residues of Hsp90s. Mitochondrial and ER Hsp90s have motifs unique to specific organisms.
The treatment of protozoan parasitic diseases is challenging, and thus identification and analysis of new drug targets is important. Parasites survive within host organisms, and some need intermediate hosts to complete their life cycle. Changing host environment puts stress on parasites, and often adaptation is accompanied by the expression of large amounts of heat shock proteins (Hsps). Among Hsps, Hsp90 proteins play an important role in stress environments. Yet, there has been little computational research on Hsp90 proteins to analyze them comparatively as potential parasitic drug targets. Here, an attempt was made to gain detailed insights into the differences between host, vector and parasitic Hsp90 proteins by large-scale bioinformatics analysis. A total of 104 Hsp90 sequences were divided into three groups based on their cellular localizations; namely cytosolic, mitochondrial and endoplasmic reticulum (ER). Further, the parasitic proteins were divided according to the type of parasite (protozoa, helminth and ectoparasite). Primary sequence analysis, phylogenetic tree calculations, motif analysis and physicochemical properties of Hsp90 proteins suggested that despite the overall structural conservation of these proteins, parasitic Hsp90 proteins have unique features which differentiate them from human ones, thus encouraging the idea that protozoan Hsp90 proteins should be further analyzed as potential drug targets.
Collapse
|
31
|
Zininga T, Makumire S, Gitau GW, Njunge JM, Pooe OJ, Klimek H, Scheurr R, Raifer H, Prinsloo E, Przyborski JM, Hoppe H, Shonhai A. Plasmodium falciparum Hop (PfHop) Interacts with the Hsp70 Chaperone in a Nucleotide-Dependent Fashion and Exhibits Ligand Selectivity. PLoS One 2015; 10:e0135326. [PMID: 26267894 PMCID: PMC4534038 DOI: 10.1371/journal.pone.0135326] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 07/22/2015] [Indexed: 12/24/2022] Open
Abstract
Heat shock proteins (Hsps) play an important role in the development and pathogenicity of malaria parasites. One of the most prominent functions of Hsps is to facilitate the folding of other proteins. Hsps are thought to play a crucial role when malaria parasites invade their host cells and during their subsequent development in hepatocytes and red blood cells. It is thought that Hsps maintain proteostasis under the unfavourable conditions that malaria parasites encounter in the host environment. Although heat shock protein 70 (Hsp70) is capable of independent folding of some proteins, its functional cooperation with heat shock protein 90 (Hsp90) facilitates folding of some proteins such as kinases and steroid hormone receptors into their fully functional forms. The cooperation of Hsp70 and Hsp90 occurs through an adaptor protein called Hsp70-Hsp90 organising protein (Hop). We previously characterised the Hop protein from Plasmodium falciparum (PfHop). We observed that the protein co-localised with the cytosol-localised chaperones, PfHsp70-1 and PfHsp90 at the blood stages of the malaria parasite. In the current study, we demonstrated that PfHop is a stress-inducible protein. We further explored the direct interaction between PfHop and PfHsp70-1 using far Western and surface plasmon resonance (SPR) analyses. The interaction of the two proteins was further validated by co-immunoprecipitation studies. We observed that PfHop and PfHsp70-1 associate in the absence and presence of either ATP or ADP. However, ADP appears to promote the association of the two proteins better than ATP. In addition, we investigated the specific interaction between PfHop TPR subdomains and PfHsp70-1/ PfHsp90, using a split-GFP approach. This method allowed us to observe that TPR1 and TPR2B subdomains of PfHop bind preferentially to the C-terminus of PfHsp70-1 compared to PfHsp90. Conversely, the TPR2A motif preferentially interacted with the C-terminus of PfHsp90. Finally, we observed that recombinant PfHop occasionally eluted as a protein species of twice its predicted size, suggesting that it may occur as a dimer. We conducted SPR analysis which suggested that PfHop is capable of self-association in presence or absence of ATP/ADP. Overall, our findings suggest that PfHop is a stress-inducible protein that directly associates with PfHsp70-1 and PfHsp90. In addition, the protein is capable of self-association. The findings suggest that PfHop serves as a module that brings these two prominent chaperones (PfHsp70-1 and PfHsp90) into a functional complex. Since PfHsp70-1 and PfHsp90 are essential for parasite growth, findings from this study are important towards the development of possible antimalarial inhibitors targeting the cooperation of these two chaperones.
Collapse
Affiliation(s)
- Tawanda Zininga
- Department of Biochemistry, School of Mathematics and Natural Sciences, University of Venda, Thohoyandou, 0950, South Africa
| | - Stanely Makumire
- Department of Biochemistry, School of Mathematics and Natural Sciences, University of Venda, Thohoyandou, 0950, South Africa
| | - Grace Wairimu Gitau
- Department of Biochemistry & Microbiology, University of Zululand, P. Bag X1001, KwaDlangezwa, 3886, South Africa
| | - James M. Njunge
- Department of Biochemistry and Microbiology, Rhodes, Grahamstown, 6140, South Africa
| | - Ofentse Jacob Pooe
- Department of Biochemistry & Microbiology, University of Zululand, P. Bag X1001, KwaDlangezwa, 3886, South Africa
| | - Hanna Klimek
- Parasitology, FB Biology, Philipps University Marburg, 35043, Marburg, Germany
| | - Robina Scheurr
- Parasitology, FB Biology, Philipps University Marburg, 35043, Marburg, Germany
| | - Hartmann Raifer
- Flow cytometry core facility, Institute for Medical Microbiology, University Clinic Marburg, Marburg, Germany
| | - Earl Prinsloo
- Biotechnology Innovation Centre, Rhodes University, Grahamstown, 6140, South Africa
| | - Jude M. Przyborski
- Parasitology, FB Biology, Philipps University Marburg, 35043, Marburg, Germany
| | - Heinrich Hoppe
- Department of Biochemistry and Microbiology, Rhodes, Grahamstown, 6140, South Africa
| | - Addmore Shonhai
- Department of Biochemistry, School of Mathematics and Natural Sciences, University of Venda, Thohoyandou, 0950, South Africa
- * E-mail:
| |
Collapse
|
32
|
Mortier J, Rakers C, Bermudez M, Murgueitio MS, Riniker S, Wolber G. The impact of molecular dynamics on drug design: applications for the characterization of ligand-macromolecule complexes. Drug Discov Today 2015; 20:686-702. [PMID: 25615716 DOI: 10.1016/j.drudis.2015.01.003] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 12/08/2014] [Accepted: 01/08/2015] [Indexed: 10/24/2022]
Abstract
Among all tools available to design new drugs, molecular dynamics (MD) simulations have become an essential technique. Initially developed to investigate molecular models with a limited number of atoms, computers now enable investigations of large macromolecular systems with a simulation time reaching the microsecond range. The reviewed articles cover four years of research to give an overview on the actual impact of MD on the current medicinal chemistry landscape with a particular emphasis on studies of ligand-protein interactions. With a special focus on studies combining computational approaches with data gained from other techniques, this review shows how deeply embedded MD simulations are in drug design strategies and articulates what the future of this technique could be.
Collapse
Affiliation(s)
- Jérémie Mortier
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Strasse 2+4, 14195 Berlin, Germany.
| | - Christin Rakers
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Strasse 2+4, 14195 Berlin, Germany
| | - Marcel Bermudez
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Strasse 2+4, 14195 Berlin, Germany
| | - Manuela S Murgueitio
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Strasse 2+4, 14195 Berlin, Germany
| | - Sereina Riniker
- Laboratory of Physical Chemistry, ETH Zürich, Vladimir-Prelog-Weg 2, CH-8093 Zurich, Switzerland
| | - Gerhard Wolber
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Strasse 2+4, 14195 Berlin, Germany.
| |
Collapse
|
33
|
The Hsp90 ensemble: coordinated Hsp90–cochaperone complexes regulate diverse cellular processes. Nat Struct Mol Biol 2014; 21:1017-21. [DOI: 10.1038/nsmb.2927] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
34
|
Brand S, Norcross NR, Thompson S, Harrison JR, Smith VC, Robinson DA, Torrie LS, McElroy SP, Hallyburton I, Norval S, Scullion P, Stojanovski L, Simeons FRC, van Aalten D, Frearson JA, Brenk R, Fairlamb AH, Ferguson MAJ, Wyatt PG, Gilbert IH, Read KD. Lead optimization of a pyrazole sulfonamide series of Trypanosoma brucei N-myristoyltransferase inhibitors: identification and evaluation of CNS penetrant compounds as potential treatments for stage 2 human African trypanosomiasis. J Med Chem 2014; 57:9855-69. [PMID: 25412409 PMCID: PMC4269550 DOI: 10.1021/jm500809c] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
![]()
Trypanosoma bruceiN-myristoyltransferase
(TbNMT) is an attractive therapeutic
target for the treatment of human African trypanosomiasis (HAT). From
previous studies, we identified pyrazole sulfonamide, DDD85646 (1), a potent inhibitor of TbNMT. Although
this compound represents an excellent lead, poor central nervous system
(CNS) exposure restricts its use to the hemolymphatic form (stage
1) of the disease. With a clear clinical need for new drug treatments
for HAT that address both the hemolymphatic and CNS stages of the
disease, a chemistry campaign was initiated to address the shortfalls
of this series. This paper describes modifications to the pyrazole
sulfonamides which markedly improved blood–brain barrier permeability,
achieved by reducing polar surface area and capping the sulfonamide.
Moreover, replacing the core aromatic with a flexible linker significantly
improved selectivity. This led to the discovery of DDD100097 (40) which demonstrated partial efficacy in a stage 2 (CNS)
mouse model of HAT.
Collapse
Affiliation(s)
- Stephen Brand
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee , Sir James Black Centre, Dundee DD1 5EH, U.K
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zierer BK, Weiwad M, Rübbelke M, Freiburger L, Fischer G, Lorenz OR, Sattler M, Richter K, Buchner J. Aktivatoren des molekularen Chaperons Hsp90 erleichtern geschwindigkeitsbestimmende Konformationsänderungen. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201406578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
36
|
Zierer BK, Weiwad M, Rübbelke M, Freiburger L, Fischer G, Lorenz OR, Sattler M, Richter K, Buchner J. Artificial accelerators of the molecular chaperone Hsp90 facilitate rate-limiting conformational transitions. Angew Chem Int Ed Engl 2014; 53:12257-62. [PMID: 25244159 DOI: 10.1002/anie.201406578] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Indexed: 11/09/2022]
Abstract
The molecular chaperone Hsp90 undergoes an ATP-driven cycle of conformational changes in which large structural rearrangements precede ATP hydrolysis. Well-established small-molecule inhibitors of Hsp90 compete with ATP-binding. We wondered whether compounds exist that can accelerate the conformational cycle. In a FRET-based screen reporting on conformational rearrangements in Hsp90 we identified compounds. We elucidated their mode of action and showed that they can overcome the intrinsic inhibition in Hsp90 which prevents these rearrangements. The mode of action is similar to that of the co-chaperone Aha1 which accelerates the Hsp90 ATPase. However, while the two identified compounds influence conformational changes, they target different aspects of the structural transitions. Also, the binding site determined by NMR spectroscopy is distinct. This study demonstrates that small molecules are capable of triggering specific rate-limiting transitions in Hsp90 by mechanisms similar to those in protein cofactors.
Collapse
Affiliation(s)
- Bettina K Zierer
- Center for Integrated Protein Science Munich (CIPSM) Department Chemie, Technische Universität München, 85747 Garching (Germany)
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Reichl MR, Braun D. Thermophoretic Manipulation of Molecules inside Living Cells. J Am Chem Soc 2014; 136:15955-60. [DOI: 10.1021/ja506169b] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Maren R. Reichl
- Systems
Biophysics, Physics
Department, NanoSystems Initiative Munich and Center for Nanoscience, Ludwig-Maximilians-Universität München, Amalienstrasse 54, 80799 München, Germany
| | - Dieter Braun
- Systems
Biophysics, Physics
Department, NanoSystems Initiative Munich and Center for Nanoscience, Ludwig-Maximilians-Universität München, Amalienstrasse 54, 80799 München, Germany
| |
Collapse
|