1
|
Olalekan SO, Obakachi VA, Badeji AA, Akinsipo (Oyelaja) OB, Familoni O, Asekun OT, Oladipo SD, Osinubi AD. Exploring the therapeutic potential of prolinamides as multi-targeted agents for Alzheimer's disease treatment: molecular docking and molecular dynamic simulation studies. In Silico Pharmacol 2024; 12:80. [PMID: 39224128 PMCID: PMC11365881 DOI: 10.1007/s40203-024-00250-z] [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: 07/03/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease (AD) presents a significant global health challenge, with its prevalence expected to rise sharply in the coming years. Despite extensive research, effective treatments addressing the multifaceted pathophysiology of AD remain elusive. This study investigates the therapeutic potential of twenty-seven prolinamides (P1 - P27), with the focus on their interactions with key proteins implicated in AD pathogenesis. Four of the compounds, namely; 10-((4-nitrophenyl)prolyl)-10 H-phenothiazine (P14), 2-((4-nitrophenyl)prolyl)isoindoline (P19), 1-(4-formylphenyl)-N-(p-tolyl)pyrrolidine-2-carboxamide (P22), and N,1-bis(4-nitrophenyl)pyrrolidine-2-carboxamide (P27) showed promising potential as Alzheimer's drug. In-silico approaches including molecular docking, molecular dynamic (MD) simulation, post md study, physicochemical and drug-likeness parameters were employed to ascertain the potential of these compounds as inhibitors of certain proteins implicated in the pathophysiology of Alzheimer's disease. Molecular docking and dynamics simulations demonstrated that P14, P19, P22 and P27 exhibited promising binding affinities towards crucial AD-associated proteins, including Beta-Secretase 1 (BACE1), Butyrylcholinesterase (BuChE), and Tau-tubulin kinase 2 (TTBK2). Structural stability analyses revealed that prolinamides, particularly P22 and P27 for BACE1 and P14 and P19 for BuChE, exhibited greater stability than their reference ligands, indicated by lower RMSD, RoG, and RMSF values. For BuChE, Rivastigmine had a docking score of -7.0 kcal/mol, a binding free energy (ΔGbind) of -22.19 ± 2.44 kcal/mol, RMSD of 1.361 ± 0.162 Å, RMSF of 9.357 ± 3.212 Å, and RoG of 22.919 ± 0.064 Å, whereas P19 exhibited a superior docking score of -10.3 kcal/mol, a significantly better ΔGbind of -33.74 ± 2.84 kcal/mol, RMSD of 1.347 ± 0.132 Å, RMSF of 8.164 ± 2.748 Å, and RoG of 22.868 ± 0.070 Å. Physicochemical and pharmacokinetic assessments affirmed the drug-likeness and bioavailability of P19 notably capable of penetrating the blood-brain barrier. Compounds P19 and P22, emerged as multi-targeted ligands, offering the potential for simultaneous modulation of multiple AD-related pathways. These findings highlight the possibilities of these compounds to be explored as novel therapeutic agents for AD. They also highlight the need for further experimental validation to confirm their efficacy and safety profiles, advancing them toward clinical application in AD management. Graphical Abstract Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00250-z.
Collapse
Affiliation(s)
- Samuel O. Olalekan
- Department of Physiology, Olabisi Onabanjo University, Sagamu Campus, Sagamu, Ogun State Nigeria
| | - Vincent A. Obakachi
- Department of Chemical Sciences, University of Johannesburg, Doornfontein Campus, P.O. Box 17011, Johannesburg, 2028 South Africa
| | - Abosede A. Badeji
- Department of Chemical Sciences, Tai Solarin University of Education, Ijagun, P.M.B. 2118, Ijebu Ode, Ogun State Nigeria
| | | | - Oluwole Familoni
- Drug Design Research Group, Department of Chemistry, University of Lagos, Akoka-Yaba, Lagos, 101245 Nigeria
| | - Olayinka T. Asekun
- Drug Design Research Group, Department of Chemistry, University of Lagos, Akoka-Yaba, Lagos, 101245 Nigeria
| | - Segun D. Oladipo
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Matieland, 7602 South Africa
- Department of Chemical Sciences, Olabisi Onabanjo University, P.M.B 2002, Ago-Iwoye, Nigeria
| | - Adejoke D. Osinubi
- Department of Chemical Sciences, Tai Solarin University of Education, Ijagun, P.M.B. 2118, Ijebu Ode, Ogun State Nigeria
- Drug Design Research Group, Department of Chemistry, University of Lagos, Akoka-Yaba, Lagos, 101245 Nigeria
| |
Collapse
|
2
|
Mousavi H, Rimaz M, Zeynizadeh B. Practical Three-Component Regioselective Synthesis of Drug-Like 3-Aryl(or heteroaryl)-5,6-dihydrobenzo[ h]cinnolines as Potential Non-Covalent Multi-Targeting Inhibitors To Combat Neurodegenerative Diseases. ACS Chem Neurosci 2024; 15:1828-1881. [PMID: 38647433 DOI: 10.1021/acschemneuro.4c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Neurodegenerative diseases (NDs) are one of the prominent health challenges facing contemporary society, and many efforts have been made to overcome and (or) control it. In this research paper, we described a practical one-pot two-step three-component reaction between 3,4-dihydronaphthalen-1(2H)-one (1), aryl(or heteroaryl)glyoxal monohydrates (2a-h), and hydrazine monohydrate (NH2NH2•H2O) for the regioselective preparation of some 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnoline derivatives (3a-h). After synthesis and characterization of the mentioned cinnolines (3a-h), the in silico multi-targeting inhibitory properties of these heterocyclic scaffolds have been investigated upon various Homo sapiens-type enzymes, including hMAO-A, hMAO-B, hAChE, hBChE, hBACE-1, hBACE-2, hNQO-1, hNQO-2, hnNOS, hiNOS, hPARP-1, hPARP-2, hLRRK-2(G2019S), hGSK-3β, hp38α MAPK, hJNK-3, hOGA, hNMDA receptor, hnSMase-2, hIDO-1, hCOMT, hLIMK-1, hLIMK-2, hRIPK-1, hUCH-L1, hPARK-7, and hDHODH, which have confirmed their functions and roles in the neurodegenerative diseases (NDs), based on molecular docking studies, and the obtained results were compared with a wide range of approved drugs and well-known (with IC50, EC50, etc.) compounds. In addition, in silico ADMET prediction analysis was performed to examine the prospective drug properties of the synthesized heterocyclic compounds (3a-h). The obtained results from the molecular docking studies and ADMET-related data demonstrated that these series of 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnolines (3a-h), especially hit ones, can really be turned into the potent core of new drugs for the treatment of neurodegenerative diseases (NDs), and/or due to the having some reactionable locations, they are able to have further organic reactions (such as cross-coupling reactions), and expansion of these compounds (for example, with using other types of aryl(or heteroaryl)glyoxal monohydrates) makes a new avenue for designing novel and efficient drugs for this purpose.
Collapse
Affiliation(s)
- Hossein Mousavi
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| | - Mehdi Rimaz
- Department of Chemistry, Payame Noor University, P.O. Box 19395-3697, Tehran 19395-3697, Iran
| | - Behzad Zeynizadeh
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| |
Collapse
|
3
|
Yeap YJ, Kandiah N, Nizetic D, Lim KL. BACE2: A Promising Neuroprotective Candidate for Alzheimer's Disease. J Alzheimers Dis 2023; 94:S159-S171. [PMID: 36463454 PMCID: PMC10473127 DOI: 10.3233/jad-220867] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2022] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia that affects millions of predominantly elderly individuals worldwide. Despite intensive research over several decades, controversies still surround the etiology of AD and the disease remains incurable. Meanwhile, new molecular players of the central amyloid cascade hypothesis have emerged and among these is a protease known as β-site APP cleavage enzyme 2 (BACE2). Unlike BACE1, BACE2 cleaves the amyloid-β protein precursor within the Aβ domain that accordingly prevents the generation of Aβ42 peptides, the aggregation of which is commonly regarded as the toxic entity that drives neurodegeneration in AD. Given this non-amyloidogenic role of BACE2, it is attractive to position BACE2 as a therapeutic target for AD. Indeed, several groups including ours have demonstrated a neuroprotective role for BACE2 in AD. In this review, we discuss emerging evidence supporting the ability of BACE2 in mitigating AD-associated pathology in various experimental systems including human pluripotent stem cell-derived cerebral organoid disease models. Alongside this, we also provide an update on the identification of single nucleotide polymorphisms occurring in the BACE2 gene that are linked to increased risk and earlier disease onset in the general population. In particular, we highlight a recently identified point mutation on BACE2 that apparently leads to sporadic early-onset AD. We believe that a better understanding of the role of BACE2 in AD would provide new insights for the development of viable therapeutic strategies for individuals with dementia.
Collapse
Affiliation(s)
- Yee Jie Yeap
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Nagaendran Kandiah
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Dean Nizetic
- Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Kah-Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Brain Sciences, Imperial College London, London, UK
- Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|
4
|
Wu C, Cui X, Sun L, Lu J, Li F, Song M, Zhang Y, Hao X, Tian C, Song M, Liu X. Aspulvinones Suppress Postprandial Hyperglycemia as Potent α-Glucosidase Inhibitors From Aspergillus terreus ASM-1. Front Chem 2021; 9:736070. [PMID: 34485249 PMCID: PMC8416056 DOI: 10.3389/fchem.2021.736070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/04/2021] [Indexed: 11/15/2022] Open
Abstract
Chemical investigation of Aspergillus terreus ASM-1 fermentation resulted in the isolation of three new prenylated aspulvinones V–X (1–3), together with the previously reported analogs, aspulvinone H (4), J-CR (5), and R (6). Their structures were elucidated by various spectroscopic methods including HRESIMS and NMR, and the absolute configurations of 2 and 3 were determined by ECD comparison. Compounds 1–6 were evaluated for α-glucosidase inhibitory effects with acarbose as positive control. As a result, compounds 1 and 4 exhibited potent α-glucosidase inhibitory activities with IC50 values of 2.2 and 4.6 µM in mixed-type manners. The thermodynamic constants recognized the interaction between inhibitors and α-glucosidase was hydrophobic force-driven spontaneous exothermic reaction. The CD spectra also indicate that the compounds 1 and 4 changed the enzyme conformation. Furthermore, compound 4 significantly suppressed the increases in postprandial blood glucose levels in the C57BL/6J mice.
Collapse
Affiliation(s)
- Changjing Wu
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, China.,College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, China.,Wuling Mountain Institute of Natural Medicine, Hubei Minzu University, Enshi, China
| | - Xiang Cui
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, China
| | - Luzhen Sun
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, China
| | - Jiajia Lu
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, China
| | - Feng Li
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, China
| | - Minghui Song
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, China
| | - Yunxia Zhang
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, China
| | - Xinqi Hao
- College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, China
| | - Congkui Tian
- Wuling Mountain Institute of Natural Medicine, Hubei Minzu University, Enshi, China
| | - Maoping Song
- College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, China
| | - Xiaomeng Liu
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, China.,College of Public Health, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
5
|
Yen YC, Kammeyer AM, Tirlangi J, Ghosh AK, Mesecar AD. A Structure-Based Discovery Platform for BACE2 and the Development of Selective BACE Inhibitors. ACS Chem Neurosci 2021; 12:581-588. [PMID: 33544569 DOI: 10.1021/acschemneuro.0c00629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The ability to perform routine structure-guided drug design for selective BACE inhibitors has been limited because of the lack of robust platform for BACE2 expression, purification, and crystallization. To overcome this limitation, we developed a platform that produces 2-3 mg of pure BACE2 protein per liter of E. coli culture, and we used this protein to design macrocyclic compounds that potently and selectively inhibit BACE1 over BACE2. Compound 2 was found to potently inhibit BACE 1 (Ki = 5 nM) with a selectivity of 214-fold over BACE2. The X-ray crystal structures of unbound BACE2 (2.2 Å) and BACE2 bound to compound 3 (3.0 Å and Ki = 7 nM) were determined and compared to the X-ray structures of BACE1 revealing the S1-S3 subsite as a selectivity determinant. This platform should enable a more rapid development of new and selective BACE inhibitors for the treatment of Alzheimer's disease or type II diabetes.
Collapse
|
6
|
Docking studies suggest the important role of interactions among the catalytic dyad and inhibitors for designing Bace1 specific inhibitors. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.127893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
7
|
Hu H, Chen Z, Xu X, Xu Y. Structure-Based Survey of the Binding Modes of BACE1 Inhibitors. ACS Chem Neurosci 2019; 10:880-889. [PMID: 30540177 DOI: 10.1021/acschemneuro.8b00420] [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] [Indexed: 12/23/2022] Open
Abstract
BACE1 is a key aspartic protease that cleaves the amyloid precursor protein to generate of the amyloid peptide that is believed to be responsible for the Alzheimer's disease amyloid cascade. It is thus recognized as a promising therapeutic target for Alzheimer's disease treatment, and large efforts have been made in the discovery of novel BACE1 inhibitors. This Review presents a systematic mining of BACE1 inhibitors based on 354 crystal structures of the BACE1 catalytic domain in complex with ligands in the Protein Data Bank. A thorough exploration on the frequency as well as the patterns of residue-ligand interactions enables us to subdivide the ligand binding pocket into 10 subsites and then identify favorable substructures of ligands for each subsite. In addition, it is found that the assembly of subsites with an 8-like shape is responsible to bind all inhibitors and four major ligand binding modes are revealed. Thus, such a systematic survey deepens our understanding of the structural requirements for establishment of BACE1-ligand interactions that determine the affinity of a ligand to BACE1, which is pivotal for structure-based lead optimization and design of novel inhibitors.
Collapse
Affiliation(s)
- Hangchen Hu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhaoqiang Chen
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yechun Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
8
|
Ghosh AK, Brindisi M, Yen YC, Lendy EK, Kovela S, Cárdenas EL, Reddy BS, Rao KV, Downs D, Huang X, Tang J, Mesecar AD. Highly Selective and Potent Human β-Secretase 2 (BACE2) Inhibitors against Type 2 Diabetes: Design, Synthesis, X-ray Structure and Structure-Activity Relationship Studies. ChemMedChem 2019; 14:545-560. [PMID: 30637955 DOI: 10.1002/cmdc.201800725] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/04/2019] [Indexed: 11/09/2022]
Abstract
Herein we present the design, synthesis, and biological evaluation of potent and highly selective β-secretase 2 (memapsin 1, beta-site amyloid precursor protein cleaving enzyme 2, or BACE 2) inhibitors. BACE2 has been recognized as an exciting new target for type 2 diabetes. The X-ray structure of BACE1 bound to inhibitor 2 a {N3 -[(1S,2R)-1-benzyl-2-hydroxy-3-[[(1S,2S)-2-hydroxy-1-(isobutylcarbamoyl)propyl]amino]propyl]-5-[methyl(methylsulfonyl)amino]-N1 -[(1R)-1-phenylpropyl]benzene-1,3-dicarboxamide} containing a hydroxyethylamine isostere was determined. Based on this structure, a computational docking study was performed which led to inhibitor 2 a-bound BACE2 models. These were used to optimize the potency and selectivity of inhibitors. A systematic structure-activity relationship study led to the identification of determinants of the inhibitors' potency and selectivity toward the BACE2 enzyme. Inhibitors 2 d [N3 -[(1S,2R)-1-benzyl-2-hydroxy-3-[[(1S,2S)-2-hydroxy-1-(isobutylcarbamoyl)pentyl]amino]propyl]-N1 -methyl-N1 -[(1R)-1-phenylpropyl]benzene-1,3-dicarboxamide; Ki =0.031 nm, selectivity over BACE1: ≈174 000-fold] and 3 l [N1 -((2S,3R)-3-hydroxy-1-phenyl-4-((3-(trifluoromethyl)benzyl)amino)butan-2-yl)-N3 ,5-dimethyl-N3 -((R)-1-phenylethyl)isophthalamide; Ki =1.6 nm, selectivity over BACE1: >500-fold] displayed outstanding potency and selectivity. Inhibitor 3 l is nonpeptide in nature and may pave the way to the development of a new class of potent and selective BACE2 inhibitors with clinical potential.
Collapse
Affiliation(s)
- Arun K Ghosh
- Department of Chemistry and Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
| | - Margherita Brindisi
- Department of Chemistry and Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
| | - Yu-Chen Yen
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Emma K Lendy
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Satish Kovela
- Department of Chemistry and Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
| | - Emilio Leal Cárdenas
- Department of Chemistry and Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
| | - Bhavanam Sekhara Reddy
- Department of Chemistry and Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
| | - Kalapala Venketeswara Rao
- Department of Chemistry and Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
| | - Deborah Downs
- Protein Studies Program, Oklahoma Medical Research Foundation, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, 73104, USA
| | - Xiangping Huang
- Protein Studies Program, Oklahoma Medical Research Foundation, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, 73104, USA
| | - Jordan Tang
- Protein Studies Program, Oklahoma Medical Research Foundation, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, 73104, USA
| | - Andrew D Mesecar
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
9
|
Xu Y, Xie L, Xie J, Liu Y, Chen W. Pelargonidin-3-O-rutinoside as a novel α-glucosidase inhibitor for improving postprandial hyperglycemia. Chem Commun (Camb) 2019; 55:39-42. [DOI: 10.1039/c8cc07985d] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Pg3R, a natural anthocyanin from strawberry, is identified as a novel α-glucosidase inhibitor and the SAR was systematically illustrated.
Collapse
Affiliation(s)
- Yang Xu
- Department of Food Science and Nutrition
- National Engineering Laboratory of Intelligent Food Technology and Equipment
- Zhejiang Key Laboratory for Agro-Food Processing
- Fuli Institute of Food Science
- Zhejiang University
| | - Lianghua Xie
- Department of Food Science and Nutrition
- National Engineering Laboratory of Intelligent Food Technology and Equipment
- Zhejiang Key Laboratory for Agro-Food Processing
- Fuli Institute of Food Science
- Zhejiang University
| | - Jiahong Xie
- Department of Food Science and Nutrition
- National Engineering Laboratory of Intelligent Food Technology and Equipment
- Zhejiang Key Laboratory for Agro-Food Processing
- Fuli Institute of Food Science
- Zhejiang University
| | - Yu Liu
- College of Life Science
- Zhejiang University
- Hangzhou 310058
- China
| | - Wei Chen
- Department of Food Science and Nutrition
- National Engineering Laboratory of Intelligent Food Technology and Equipment
- Zhejiang Key Laboratory for Agro-Food Processing
- Fuli Institute of Food Science
- Zhejiang University
| |
Collapse
|
10
|
Zogota R, Kinena L, Withers-Martinez C, Blackman MJ, Bobrovs R, Pantelejevs T, Kanepe-Lapsa I, Ozola V, Jaudzems K, Suna E, Jirgensons A. Peptidomimetic plasmepsin inhibitors with potent anti-malarial activity and selectivity against cathepsin D. Eur J Med Chem 2018; 163:344-352. [PMID: 30529637 PMCID: PMC6336538 DOI: 10.1016/j.ejmech.2018.11.068] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/26/2018] [Accepted: 11/28/2018] [Indexed: 11/18/2022]
Abstract
Following up the open initiative of anti-malarial drug discovery, a GlaxoSmithKline (GSK) phenotypic screening hit was developed to generate hydroxyethylamine based plasmepsin (Plm) inhibitors exhibiting growth inhibition of the malaria parasite Plasmodium falciparum at nanomolar concentrations. Lead optimization studies were performed with the aim of improving Plm inhibition selectivity versus the related human aspartic protease cathepsin D (Cat D). Optimization studies were performed using Plm IV as a readily accessible model protein, the inhibition of which correlates with anti-malarial activity. Guided by sequence alignment of Plms and Cat D, selectivity-inducing structural motifs were modified in the S3 and S4 sub-pocket occupying substituents of the hydroxyethylamine inhibitors. This resulted in potent anti-malarials with an up to 50-fold Plm IV/Cat D selectivity factor. More detailed investigation of the mechanism of action of the selected compounds revealed that they inhibit maturation of the P. falciparum subtilisin-like protease SUB1, and also inhibit parasite egress from erythrocytes. Our results indicate that the anti-malarial activity of the compounds is linked to inhibition of the SUB1 maturase plasmepsin subtype Plm X. Peptidomimimetic plasmepsin inhibitors are developed using Plm IV as a model enzyme. Up to 50-fold selectivity against Cathepsin D is reached. Compounds show growth inhibition of P. falciparum at nanomolar concentrations. Inhibition of SUB1 maturation and parasite egress imply (co)inhibition of Plm X.
Collapse
Affiliation(s)
- Rimants Zogota
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | - Linda Kinena
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | | | - Michael J Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK; Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Raitis Bobrovs
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | - Teodors Pantelejevs
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | - Iveta Kanepe-Lapsa
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | - Vita Ozola
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | - Kristaps Jaudzems
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | - Edgars Suna
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia.
| | - Aigars Jirgensons
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia.
| |
Collapse
|
11
|
Graham SE, Leja N, Carlson HA. MixMD Probeview: Robust Binding Site Prediction from Cosolvent Simulations. J Chem Inf Model 2018; 58:1426-1433. [PMID: 29905479 DOI: 10.1021/acs.jcim.8b00265] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mixed-solvent molecular dynamics (MixMD) is a cosolvent simulation technique for identifying binding hotspots and specific favorable interactions on a protein's surface. MixMD studies have the ability to identify these biologically relevant sites by examining the occupancy of the cosolvent over the course of the simulation. However, previous MixMD analysis required a great deal of manual inspection to identify relevant sites. To address this limitation, we have developed MixMD Probeview as a plugin for the freely available, open-source version of the molecular visualization program PyMOL. MixMD Probeview incorporates two analysis procedures: (1) identifying and ranking whole binding sites and (2) identifying and ranking local maxima for each probe type. These functionalities were validated using four common benchmark proteins, including two with both active and allosteric sites. In addition, three different cosolvent procedures were compared to examine the impact of including more than one cosolvent in the simulations. For all systems tested, MixMD Probeview successfully identified known active and allosteric sites based on the total occupancy of neutral probe molecules. As an easy-to-use PyMOL plugin, we expect that MixMD Probeview will facilitate identification and analysis of binding sites from cosolvent simulations performed on a wide range of systems.
Collapse
|
12
|
Voytyuk I, De Strooper B, Chávez-Gutiérrez L. Modulation of γ- and β-Secretases as Early Prevention Against Alzheimer's Disease. Biol Psychiatry 2018; 83:320-327. [PMID: 28918941 DOI: 10.1016/j.biopsych.2017.08.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 08/01/2017] [Accepted: 08/03/2017] [Indexed: 01/18/2023]
Abstract
The genetic evidence implicating amyloid-β in the initial stage of Alzheimer's disease is unequivocal. However, the long biochemical and cellular prodromal phases of the disease suggest that dementia is the result of a series of molecular and cellular cascades whose nature and connections remain unknown. Therefore, it is unlikely that treatments directed at amyloid-β will have major clinical effects in the later stages of the disease. We discuss the two major candidate therapeutic targets to lower amyloid-β in a preventive mode, i.e., γ- and β-secretase; the rationale behind these two targets; and the current state of the field.
Collapse
Affiliation(s)
- Iryna Voytyuk
- KU Leuven Department for Neurosciences, Leuven, Belgium; VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | - Bart De Strooper
- KU Leuven Department for Neurosciences, Leuven, Belgium; VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium; UK Dementia Research Institute, University College, London, United Kingdom.
| | - Lucía Chávez-Gutiérrez
- KU Leuven Department for Neurosciences, Leuven, Belgium; VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| |
Collapse
|
13
|
Um J, Lee JH, Jung DW, Williams DR. Re-education begins at home: an overview of the discovery of in vivo-active small molecule modulators of endogenous stem cells. Expert Opin Drug Discov 2018; 13:307-326. [PMID: 29421943 DOI: 10.1080/17460441.2018.1437140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Degenerative diseases, such as Alzheimer's disease, heart disease and arthritis cause great suffering and are major socioeconomic burdens. An attractive treatment approach is stem cell transplantation to regenerate damaged or destroyed tissues. However, this can be problematic. For example, donor cells may not functionally integrate into the host tissue. An alternative methodology is to deliver bioactive agents, such as small molecules, directly into the diseased tissue to enhance the regenerative potential of endogenous stem cells. Areas covered: In this review, the authors discuss the necessity of developing these small molecules to treat degenerative diseases and survey progress in their application as therapeutics. They describe both the successes and caveats of developing small molecules that target endogenous stem cells to induce tissue regeneration. This article is based on literature searches which encompass databases for biomedical research and clinical trials. These small molecules are also categorized per their target disease and mechanism of action. Expert opinion: The development of small molecules targeting endogenous stem cells is a high-profile research area. Some compounds have made the successful transition to the clinic. Novel approaches, such as modulating the stem cell niche or targeted delivery to disease sites, should increase the likelihood of future successes in this field.
Collapse
Affiliation(s)
- JungIn Um
- a New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology , Buk-Gu , Gwangju , Republic of Korea
| | - Ji-Hyung Lee
- a New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology , Buk-Gu , Gwangju , Republic of Korea
| | - Da-Woon Jung
- a New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology , Buk-Gu , Gwangju , Republic of Korea
| | - Darren R Williams
- a New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology , Buk-Gu , Gwangju , Republic of Korea
| |
Collapse
|