1
|
Dechenne J, Wierzbicka M, Krimou R, El Aakchioui A, Malo Pueyo J, Messens J, Fillet M, Spillier Q, Frédérick R. Examining Arginase-1 Trimerization Uncovers a Promising Allosteric Site for Inhibition. J Med Chem 2025; 68:1433-1445. [PMID: 39748145 DOI: 10.1021/acs.jmedchem.4c01993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Arginase-1 (ARG-1) is a promising target for cancer immunotherapy, but the small size and the highly polar nature of its catalytic site present significant challenges for inhibitor development. An alternative strategy to induce enzyme inhibition by targeting protein oligomerization has been developed recently, offering several advantages such as increased selectivity, promotion of protein degradation, and potential substoichiometric inhibition. In this study, we demonstrated that only trimeric ARG-1 is active, which was confirmed by producing monomeric arginase-1. Through in silico-driven site-directed mutagenesis, we identified an allosteric site involving five key amino acids responsible for ARG-1 trimerization. We further demonstrated the covalent modification of a key arginine residue within this pocket using phenylglyoxal disrupted ARG-1 oligomerization. Although phenylglyoxal has limited potency, it effectively supports the concept of ARG-1 inhibition via homomeric disruption, validating this allosteric targeting approach.
Collapse
Affiliation(s)
- Juhans Dechenne
- Louvain Drug Research Institute (LDRI), Medicinal Chemistry Research Group (CMFA), Université Catholique de Louvain (UCLouvain), Brussels B-1200, Belgium
| | - Magdalena Wierzbicka
- Laboratory for the Analysis of Medicines (CIRM), Université de Liège (ULG), Liège B-4000, Belgium
| | - Reda Krimou
- Louvain Drug Research Institute (LDRI), Medicinal Chemistry Research Group (CMFA), Université Catholique de Louvain (UCLouvain), Brussels B-1200, Belgium
| | - Asia El Aakchioui
- Louvain Drug Research Institute (LDRI), Medicinal Chemistry Research Group (CMFA), Université Catholique de Louvain (UCLouvain), Brussels B-1200, Belgium
| | - Julia Malo Pueyo
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, Brussels B-1050, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, Brussels B-1050, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels B-1050, Belgium
| | - Joris Messens
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, Brussels B-1050, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, Brussels B-1050, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels B-1050, Belgium
| | - Marianne Fillet
- Laboratory for the Analysis of Medicines (CIRM), Université de Liège (ULG), Liège B-4000, Belgium
| | - Quentin Spillier
- Louvain Drug Research Institute (LDRI), Medicinal Chemistry Research Group (CMFA), Université Catholique de Louvain (UCLouvain), Brussels B-1200, Belgium
| | - Raphaël Frédérick
- Louvain Drug Research Institute (LDRI), Medicinal Chemistry Research Group (CMFA), Université Catholique de Louvain (UCLouvain), Brussels B-1200, Belgium
| |
Collapse
|
2
|
Sherry D, Sayed Y. Unveiling a Hidden Pocket in HIV-1 Protease: New Insights Into Retroviral Protease Cantilever-Tip Region Characteristics. Proteins 2024; 92:1398-1412. [PMID: 39109919 DOI: 10.1002/prot.26735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 11/08/2024]
Abstract
The HIV-1 protease is critical for the process of viral maturation and as such, it is one of the most well characterized proteins in the Protein Data Bank. There is some evidence to suggest that the HIV-1 protease is capable of accommodating small molecule fragments at several locations on its surface outside of the active site. However, some pockets on the surface of proteins remain unformed in the apo structure and are termed "cryptic sites." To date, no cryptic sites have been identified in the structure of HIV-1 protease. Here, we characterize a novel cryptic cantilever pocket on the surface of the HIV-1 protease through mixed-solvent molecular dynamics simulations using several probes. Interestingly, we noted that several homologous retroviral proteases exhibit evolutionarily conserved dynamics in the cantilever region and possess a conserved pocket in the cantilever region. Immobilization of the cantilever region of the HIV-1 protease via disulfide cross-linking resulted in curling-in of the flap tips and the propensity for the protease to adopt a semi-open flap conformation. Structure-based analysis and fragment-based screening of the cryptic cantilever pocket suggested that the pocket may be capable of accommodating ligand structures. Furthermore, molecular dynamics simulations of a top scoring fragment bound to the cryptic pocket illustrated altered flap dynamics of the fragment-bound enzyme. Together, these results suggest that the mobility of the cantilever region plays a key role in the global dynamics of retroviral proteases. Therefore, the cryptic cantilever pocket of the HIV-1 protease may represent an interesting target for future in vitro studies.
Collapse
Affiliation(s)
- Dean Sherry
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, South Africa
| | - Yasien Sayed
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
3
|
Mauchauffée E, Leroy J, Chamcham J, Ejjoummany A, Maurel M, Nauton L, Ramassamy B, Mezghenna K, Boucher JL, Lajoix AD, Hernandez JF. S-Ethyl-Isothiocitrullin-Based Dipeptides and 1,2,4-Oxadiazole Pseudo-Dipeptides: Solid Phase Synthesis and Evaluation as NO Synthase Inhibitors. Molecules 2023; 28:5085. [PMID: 37446746 DOI: 10.3390/molecules28135085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
We previously reported dipeptidomimetic compounds as inhibitors of neuronal and/or inducible NO synthases (n/iNOS) with significant selectivity against endothelial NOS (eNOS). They were composed of an S-ethylisothiocitrullin-like moiety linked to an extension through a peptide bond or a 1,2,4-oxadiazole link. Here, we developed two further series where the extension size was increased to establish more favorable interactions in the NOS substrate access channel. The extension was introduced on the solid phase by the reductive alkylation of an amino-piperidine moiety or an aminoethyl segment in the case of dipeptide-like and 1,2,4-oxadiazole compounds, respectively, with various benzaldehydes. Compared to the previous series, more potent inhibitors were identified with IC50 in the micromolar to the submicromolar range, with significant selectivity toward nNOS. As expected, most compounds did not inhibit eNOS, and molecular modeling was carried out to characterize the reasons for the selectivity toward nNOS over eNOS. Spectral studies showed that compounds were interacting at the heme active site. Finally, selected inhibitors were found to inhibit intra-cellular iNOS and nNOS expressed in RAW264.7 and INS-1 cells, respectively.
Collapse
Affiliation(s)
- Elodie Mauchauffée
- Institut des Biomolécules Max Mousseron, CNRS, Univ. Montpellier, ENSCM, Pôle Chimie Balard, 34293 Montpellier, France
| | - Jérémy Leroy
- Centre Biocommunication en Cardio-Métabolique, Univ. Montpellier, UFR Pharmacie, 34093 Montpellier, France
| | - Jihanne Chamcham
- Institut des Biomolécules Max Mousseron, CNRS, Univ. Montpellier, ENSCM, Pôle Chimie Balard, 34293 Montpellier, France
| | - Abdelaziz Ejjoummany
- Institut des Biomolécules Max Mousseron, CNRS, Univ. Montpellier, ENSCM, Pôle Chimie Balard, 34293 Montpellier, France
| | - Manon Maurel
- Institut des Biomolécules Max Mousseron, CNRS, Univ. Montpellier, ENSCM, Pôle Chimie Balard, 34293 Montpellier, France
| | - Lionel Nauton
- Institut de Chimie de Clermont-Ferrand, Université Clermont-Auvergne, CNRS, 63178 Aubière, France
| | - Booma Ramassamy
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, UMR 8601, CNRS, Université Paris Descartes, CEDEX 06, 75270 Paris, France
| | - Karima Mezghenna
- Centre Biocommunication en Cardio-Métabolique, Univ. Montpellier, UFR Pharmacie, 34093 Montpellier, France
| | - Jean-Luc Boucher
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, UMR 8601, CNRS, Université Paris Descartes, CEDEX 06, 75270 Paris, France
| | - Anne-Dominique Lajoix
- Centre Biocommunication en Cardio-Métabolique, Univ. Montpellier, UFR Pharmacie, 34093 Montpellier, France
| | - Jean-François Hernandez
- Institut des Biomolécules Max Mousseron, CNRS, Univ. Montpellier, ENSCM, Pôle Chimie Balard, 34293 Montpellier, France
| |
Collapse
|
4
|
Rehman AU, Khurshid B, Ali Y, Rasheed S, Wadood A, Ng HL, Chen HF, Wei Z, Luo R, Zhang J. Computational approaches for the design of modulators targeting protein-protein interactions. Expert Opin Drug Discov 2023; 18:315-333. [PMID: 36715303 PMCID: PMC10149343 DOI: 10.1080/17460441.2023.2171396] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 01/18/2023] [Indexed: 01/31/2023]
Abstract
BACKGROUND Protein-protein interactions (PPIs) are intriguing targets for designing novel small-molecule inhibitors. The role of PPIs in various infectious and neurodegenerative disorders makes them potential therapeutic targets . Despite being portrayed as undruggable targets, due to their flat surfaces, disorderedness, and lack of grooves. Recent progresses in computational biology have led researchers to reconsider PPIs in drug discovery. AREAS COVERED In this review, we introduce in-silico methods used to identify PPI interfaces and present an in-depth overview of various computational methodologies that are successfully applied to annotate the PPIs. We also discuss several successful case studies that use computational tools to understand PPIs modulation and their key roles in various physiological processes. EXPERT OPINION Computational methods face challenges due to the inherent flexibility of proteins, which makes them expensive, and result in the use of rigid models. This problem becomes more significant in PPIs due to their flexible and flat interfaces. Computational methods like molecular dynamics (MD) simulation and machine learning can integrate the chemical structure data into biochemical and can be used for target identification and modulation. These computational methodologies have been crucial in understanding the structure of PPIs, designing PPI modulators, discovering new drug targets, and predicting treatment outcomes.
Collapse
Affiliation(s)
- Ashfaq Ur Rehman
- Departments of Molecular Biology and Biochemistry, Chemical and Biomolecular Engineering, Materials Science and Engineering, and Biomedical Engineering, Graduate Program in Chemical and Materials Physics, University of California Irvine, Irvine, California, USA
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai, Zhejiang, China
| | - Beenish Khurshid
- Department of Biochemistry, Abdul Wali Khan University Mardan, Pakistan
| | - Yasir Ali
- National Center for Bioinformatics, Quaid-e-Azam University, Islamabad, Pakistan
| | - Salman Rasheed
- National Center for Bioinformatics, Quaid-e-Azam University, Islamabad, Pakistan
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Pakistan
| | - Ho-Leung Ng
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas, USA
| | - Hai-Feng Chen
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, Zhejiang, China
| | - Zhiqiang Wei
- Medicinal Chemistry and Bioinformatics Center, Ocean University of China, Qingdao, Shandong, China
| | - Ray Luo
- Departments of Molecular Biology and Biochemistry, Chemical and Biomolecular Engineering, Materials Science and Engineering, and Biomedical Engineering, Graduate Program in Chemical and Materials Physics, University of California Irvine, Irvine, California, USA
| | - Jian Zhang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai, Zhejiang, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Chenicheri S, Ramachandran R, Rajamanikam U. Antimicrobial effects of hydroxyapatite mosaicked polyvinyl alcohol-alginate semi-interpenetrating hydrogel-loaded with ethanolic extract of Glycyrrhiza glabra against oral pathogens. Prog Biomater 2022; 11:373-383. [PMID: 35969367 DOI: 10.1007/s40204-022-00199-2] [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: 03/23/2022] [Accepted: 07/27/2022] [Indexed: 11/25/2022] Open
Abstract
Glycyrrhiza glabra (GG) elicits protective effects against periodontal diseases. However, the sustained bioavailability of GG extract at therapeutic concentration warrants ideal delivery vehicles. Present study has focused on the design, fabrication, and evaluations of ethanolic-crude extract of GG-loaded semi-interpenetrating network (semi-IPN) hydrogel (HAAPS-GG) using alginic acid and polyvinyl alcohol (PVA) hydrogel mosaicked with HA for periodontal regeneration. The study has examined the performance of the hydrogel against the selected oral pathogens S. mutans, E. faecalis, L. acidophilus and C. albicans. HAAPS-GG was successfully fabricated and the surface functional groups were confirmed by attenuated total reflectance-infrared (ATR-IR) spectroscopy. HAAPS-GG displayed interconnecting pores, hydrophilicity and excellent water profile contributing to the biocompatibility as evident from direct contact and MTT assay in L929 fibroblasts. The hydrogel was mechanically stable and was immunocompatible owing to the relatively decreased levels of pro-inflammatory mediators COX2, 5LPO, iNOS and MPO in RAW 264.7 macrophages. In addition, the transcript analysis on RAW 264.7 revealed the down-regulation of inflammatory transcription factor NF-κβ and the pro-inflammatory cytokine TNF-α. Importantly, HAAPS-GG arrested the progression of periodontal pathogens predominantly S. mutans, and C. albicans as evident by disc diffusion assay, MTT assay and confocal microscopy. Overall, the HAAPS-GG system offers promising translational avenues in periodontal regeneration.
Collapse
Affiliation(s)
- Smitha Chenicheri
- Department of Microbiology, PMS College of Dental Science and Research, Thiruvanathapuram, 695028, Kerala, India.
- Biomaterial Divisions, Centre for Research in Molecular and Applied Sciences (CRMAS), Thiruvanathapuram, 695006, Kerala, India.
| | - Rajesh Ramachandran
- Biomaterial Divisions, Centre for Research in Molecular and Applied Sciences (CRMAS), Thiruvanathapuram, 695006, Kerala, India
| | - Usha Rajamanikam
- Karpagam Academy for Higher Education, Coimbatore, 641021, Tamilnadu, India
| |
Collapse
|
6
|
Uelisson da Silva T, Tomaz da Silva E, de Carvalho Pougy K, Henrique da Silva Lima C, de Paula Machado S. Molecular modeling of indazole-3-carboxylic acid and its metal complexes (Zn, Ni, Co, Fe and Mn) as NO synthase inhibitors: DFT calculations, docking studies and molecular dynamics simulations. INORG CHEM COMMUN 2022. [DOI: 10.1016/j.inoche.2021.109120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
7
|
Zhu LQ, Fan XH, Li JF, Chen JH, Liang Y, Hu XL, Ma SM, Hao XY, Shi T, Wang Z. Discovery of a novel inhibitor of nitric oxide production with potential therapeutic effect on acute inflammation. Bioorg Med Chem Lett 2021; 44:128106. [PMID: 33991630 DOI: 10.1016/j.bmcl.2021.128106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/28/2021] [Accepted: 05/08/2021] [Indexed: 10/21/2022]
Abstract
Inflammation as a host's excessive immune response to stimulation, is involved in the development of numerous diseases. To discover novel anti-inflammatory agents and based on our previous synthetic work on marine natural product Chrysamide B, it and a series of derivatives were synthesized and evaluated for their anti-inflammatory activity on inhibition of LPS-induced NO production. Then the preliminary structure-activity relationships were conducted. Among them, Chrysamide B is the most potent anti-inflammatory agent with low cytotoxicity and strong inhibition on the production of NO (IC50 = 0.010 μM) and the activity of iNOS (IC50 = 0.082 μM) in LPS-stimulated RAW 264.7 cells. Primary studies suggested that the mechanism of action may be that it interfered the formation of active dimeric iNOS but not affected transcription and translation. Furthermore, its good performance of anti-inflammatory effect on LPS-induced multiple inflammatory cytokines production, carrageenan-induced paw edema, and endotoxin-induced septic mice, was observed. We believe that these findings would provide an idea for the further modification and research of these analogs in the future.
Collapse
Affiliation(s)
- Long-Qing Zhu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Xiao-Hong Fan
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jun-Fang Li
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jin-Hong Chen
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yan Liang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Xiao-Ling Hu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Shu-Meng Ma
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Xiang-Yong Hao
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Tao Shi
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| | - Zhen Wang
- School of Pharmaceutical Science, University of South China, Hengyang 421001, China; School of Pharmacy, Lanzhou University, Lanzhou 730000, China; State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
8
|
Zhu HY, Hong FF, Yang SL. The Roles of Nitric Oxide Synthase/Nitric Oxide Pathway in the Pathology of Vascular Dementia and Related Therapeutic Approaches. Int J Mol Sci 2021; 22:ijms22094540. [PMID: 33926146 PMCID: PMC8123648 DOI: 10.3390/ijms22094540] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022] Open
Abstract
Vascular dementia (VaD) is the second most common form of dementia worldwide. It is caused by cerebrovascular disease, and patients often show severe impairments of advanced cognitive abilities. Nitric oxide synthase (NOS) and nitric oxide (NO) play vital roles in the pathogenesis of VaD. The functions of NO are determined by its concentration and bioavailability, which are regulated by NOS activity. The activities of different NOS subtypes in the brain are partitioned. Pathologically, endothelial NOS is inactivated, which causes insufficient NO production and aggravates oxidative stress before inducing cerebrovascular endothelial dysfunction, while neuronal NOS is overactive and can produce excessive NO to cause neurotoxicity. Meanwhile, inflammation stimulates the massive expression of inducible NOS, which also produces excessive NO and then induces neuroinflammation. The vicious circle of these kinds of damage having impacts on each other finally leads to VaD. This review summarizes the roles of the NOS/NO pathway in the pathology of VaD and also proposes some potential therapeutic methods that target this pathway in the hope of inspiring novel ideas for VaD therapeutic approaches.
Collapse
Affiliation(s)
- Han-Yan Zhu
- Department of Physiology, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China;
- Queen Marry College, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China
| | - Fen-Fang Hong
- Teaching Center, Department of Experimental, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China
- Correspondence: (F.-F.H.); (S.-L.Y.)
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China;
- Correspondence: (F.-F.H.); (S.-L.Y.)
| |
Collapse
|
9
|
Targeting protein self-association in drug design. Drug Discov Today 2021; 26:1148-1163. [PMID: 33548462 DOI: 10.1016/j.drudis.2021.01.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/24/2020] [Accepted: 01/26/2021] [Indexed: 01/05/2023]
Abstract
Protein self-association is a universal phenomenon essential for stability and molecular recognition. Disrupting constitutive homomers constitutes an original and emerging strategy in drug design. Inhibition of homomeric proteins can be achieved through direct complex disruption, subunit intercalation, or by promoting inactive oligomeric states. Targeting self-interaction grants several advantages over active site inhibition because of the stimulation of protein degradation, the enhancement of selectivity, substoichiometric inhibition, and by-pass of compensatory mechanisms. This new landscape in protein inhibition is driven by the development of biophysical and biochemical tools suited for the study of homomeric proteins, such as differential scanning fluorimetry (DSF), native mass spectrometry (MS), Förster resonance energy transfer (FRET) spectroscopy, 2D nuclear magnetic resonance (NMR), and X-ray crystallography. In this review, we discuss the different aspects of this new paradigm in drug design.
Collapse
|
10
|
Chen LZ, Shu HY, Wu J, Yu YL, Ma D, Huang X, Liu MM, Liu XH, Shi JB. Discovery and development of novel pyrimidine and pyrazolo/thieno-fused pyrimidine derivatives as potent and orally active inducible nitric oxide synthase dimerization inhibitor with efficacy for arthritis. Eur J Med Chem 2021; 213:113174. [PMID: 33515864 DOI: 10.1016/j.ejmech.2021.113174] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/24/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022]
Abstract
In order to discover and develop drug-like anti-inflammatory agents against arthritis, based on "Hit" we found earlier and to overcome drawbacks of toxicity, twelve series of total 89 novel pyrimidine, pyrazolo[4,3-d]pyrimidine and thieno[3,2-d]pyrimidine derivatives were designed, synthesized and screened for their anti-inflammatory activity against NO and toxicity for normal liver cells (LO2). Relationships of balance toxicity and activity have been summarized through multi-steps, and title compounds 22o, 22l were found to show lower toxicity (against LO2: IC50 = 2934, 2301 μM, respectively) and potent effect against NO release (IR = 98.3, 97.67%, at 10 μM, respectively). Furthermore, compound 22o showed potent iNOS inhibitory activity with value of IC50 is 0.96 μM and could interfere stability and formation of the active dimeric iNOS. It's anti-inflammatory activity in vivo was assessed by AIA rat model. Furthermore, the results of metabolic stability, CYP, PK study in vivo, acute toxicity study and subacute toxicity assessment indicated this compound had good drug-like properties for treatment.
Collapse
Affiliation(s)
- Liu Zeng Chen
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Hai Yang Shu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Jing Wu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Yun Long Yu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Duo Ma
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Xin Huang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Ming Ming Liu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Xin Hua Liu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China.
| | - Jing Bo Shi
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China.
| |
Collapse
|
11
|
Mittal A, Kakkar R. Nitric Oxide Synthases and Their Inhibitors: A Review. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180816666190222154457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nitric Oxide (NO), an important biological mediator, is involved in the regulation of the cardiovascular, nervous and immune systems in mammals. Synthesis of NO is catalyzed by its biosynthetic enzyme, Nitric Oxide Synthase (NOS). There are three main isoforms of the enzyme, neuronal NOS, endothelial NOS and inducible NOS, which have very similar structures but differ in their expression and activities. NO is produced in the active site of the enzyme in two distinct cycles from oxidation of the substrate L-arg (L-arginine) in nicotinamide adenine dinucleotide phosphate (NADPH)-dependent reaction. NOS has gained considerable attention of biochemists due to its complexity and unique catalytic mechanism. The review focuses on NOS structure, its function and catalytic reaction mechanism. In particular, the review is concluded with a discussion on the role of all three isoforms of NOS in physiological and pathological conditions and their inhibitors with a focus on the role of computational techniques in their development.
Collapse
Affiliation(s)
- Anshika Mittal
- Computational Chemistry Laboratory, Department of Chemistry, University of Delhi, Delhi-110007, India
| | - Rita Kakkar
- Computational Chemistry Laboratory, Department of Chemistry, University of Delhi, Delhi-110007, India
| |
Collapse
|
12
|
Touati-Jallabe Y, Tintillier T, Mauchauffée E, Boucher JL, Leroy J, Ramassamy B, Hamzé A, Mezghenna K, Bouzekrini A, Verna C, Martinez J, Lajoix AD, Hernandez JF. Solid-Phase Synthesis of Substrate-Based Dipeptides and Heterocyclic Pseudo-dipeptides as Potential NO Synthase Inhibitors. ChemMedChem 2020; 15:517-531. [PMID: 32027778 DOI: 10.1002/cmdc.201900659] [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: 11/26/2019] [Revised: 02/03/2020] [Indexed: 11/06/2022]
Abstract
More than 160 arginine analogues modified on the C-terminus via either an amide bond or a heterocyclic moiety (1,2,4-oxadiazole, 1,3,4-oxadiazole and 1,2,4-triazole) were prepared as potential inhibitors of NO synthases (NOS). A methodology involving formation of a thiocitrulline intermediate linked through its side-chain on a solid support followed by modification of its carboxylate group was developed. Finally, the side-chain thiourea group was either let unchanged, S-alkylated (Me, Et) or guanidinylated (Me, Et) to yield respectively after TFA treatment the corresponding thiocitrulline, S-Me/Et-isothiocitrulline and N-Me/Et-arginine substrate analogues. They all were tested against three recombinant NOS isoforms. Several compounds containing a S-Et- or a S-Me-Itc moiety and mainly belonging to both the dipeptide-like and 1,2,4-oxadiazole series were shown to inhibit nNOS and iNOS with IC50 in the 1-50 μM range. Spectral studies confirmed that these new compounds interacted at the heme active site. The more active compounds were found to inhibit intra-cellular iNOS expressed in RAW264.7 and INS-1 cells with similar efficiency than the reference compounds L-NIL and SEIT.
Collapse
Affiliation(s)
- Youness Touati-Jallabe
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France.,Avara Pharmaceutical Services, Boucherville, QC, J4B 7 K8, Canada
| | - Thibault Tintillier
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France.,Asymptote Project Management, 1 rue Edisson, 69500, Bron, France
| | - Elodie Mauchauffée
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France
| | - Jean-Luc Boucher
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques UMR8601, CNRS, Université Paris-Descartes, 45 rue des Saints Pères, 75270, Paris Cedex 06, France
| | - Jérémy Leroy
- Centre Biocommunication en Cardio-métabolique, Université Montpellier, Faculté de Pharmacie, 34000, Montpellier, France
| | - Booma Ramassamy
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques UMR8601, CNRS, Université Paris-Descartes, 45 rue des Saints Pères, 75270, Paris Cedex 06, France
| | - Abdallah Hamzé
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France.,Current address: BioCIS, UMR 8076, CNRS, Université Paris Sud, Université Paris-Saclay, 92296, Châtenay-Malabry, France
| | - Karima Mezghenna
- Centre Biocommunication en Cardio-métabolique, Université Montpellier, Faculté de Pharmacie, 34000, Montpellier, France
| | - Amine Bouzekrini
- Centre Biocommunication en Cardio-métabolique, Université Montpellier, Faculté de Pharmacie, 34000, Montpellier, France
| | - Claudia Verna
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France
| | - Jean Martinez
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France
| | - Anne-Dominique Lajoix
- Centre Biocommunication en Cardio-métabolique, Université Montpellier, Faculté de Pharmacie, 34000, Montpellier, France
| | - Jean-François Hernandez
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France
| |
Collapse
|
13
|
Small molecule inhibitors and stimulators of inducible nitric oxide synthase in cancer cells from natural origin (phytochemicals, marine compounds, antibiotics). Biochem Pharmacol 2020; 176:113792. [PMID: 31926145 DOI: 10.1016/j.bcp.2020.113792] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 01/03/2020] [Indexed: 02/06/2023]
Abstract
Nitric oxide synthases (NOS) are a family of isoforms, which generate nitric oxide (NO). NO is one of the smallest molecules in nature and acts mainly as a potent vasodilator. It participates in various biological processes ranging from physiological to pathological conditions. Inducible NOS (iNOS, NOS2) is a calcium-independent and inducible isoform. Despite high iNOS expression in many tumors, the role of iNOS is still unclear and complex with both enhancing and prohibiting actions in tumorigenesis. Nature presents a broad variety of natural stimulators and inhibitors, which may either promote or inhibit iNOS response. In the present review, we give an overview of iNOS-modulating agents with a special focus on both natural and synthetic molecules and their effects in related biological processes. The role of iNOS in physiological and pathological conditions is also discussed.
Collapse
|
14
|
Rodrigues RP, Ardisson JS, Ribeiro Gonçalves RDC, Oliveira TB, Barreto da Silva V, Kawano DF, Kitagawa RR. Search for Potential Inducible Nitric Oxide Synthase Inhibitors with Favorable ADMET Profiles for the Therapy of Helicobacter pylori Infections. Curr Top Med Chem 2020; 19:2795-2804. [DOI: 10.2174/1568026619666191112105650] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/04/2019] [Accepted: 10/04/2019] [Indexed: 11/22/2022]
Abstract
Background:
Helicobacter pylori is a gram-negative bacterium related to chronic gastritis, peptic
ulcer and gastric carcinoma. During its infection process, promotes excessive inflammatory response, increasing
the release of reactive species and inducing the production of pro-inflammatory mediators. Inducible Nitric
Oxide Synthase (iNOS) plays a crucial role in the gastric carcinogenesis process and a key mediator of inflammation
and host defense systems, which is expressed in macrophages induced by inflammatory stimuli. In
chronic diseases such as Helicobacter pylori infections, the overproduction of NO due to the prolonged induction
of iNOS is of major concern.
Objective:
In this sense, the search for potential iNOS inhibitors is a valuable strategy in the overall process
of Helicobacter pylori pathogeny.
Method:
In silico techniques were applied in the search of interesting compounds against Inducible Nitric
Oxide Synthase enzyme in a chemical space of natural products and derivatives from the Analyticon Discovery
databases.
Results:
The five compounds with the best iNOS inhibition profile were selected for activity and toxicity predictions.
Compound 9 (CAS 88198-99-6) displayed significant potential for iNOS inhibition, forming hydrogen
bonds with residues from the active site and an ionic interaction with heme. This compound also displayed
good bioavailability and absence of toxicity/or from its probable metabolites.
Conclusion:
The top-ranked compounds from the virtual screening workflow show promising results regarding
the iNOS inhibition profile. The results evidenced the importance of the ionic bonding during docking selection,
playing a crucial role in binding and positioning during ligand-target selection for iNOS.
Collapse
Affiliation(s)
- Ricardo Pereira Rodrigues
- Graduate Program in Pharmaceutical Sciences, Health Sciences Center - CCS, Federal University of Espirito Santo - UFES, Marechal Campos Av., 1468, Vitoria 29043-900, ES, Brazil
| | - Juliana Santa Ardisson
- Graduate Program in Pharmaceutical Sciences, Health Sciences Center - CCS, Federal University of Espirito Santo - UFES, Marechal Campos Av., 1468, Vitoria 29043-900, ES, Brazil
| | - Rita de Cássia Ribeiro Gonçalves
- Graduate Program in Pharmaceutical Sciences, Health Sciences Center - CCS, Federal University of Espirito Santo - UFES, Marechal Campos Av., 1468, Vitoria 29043-900, ES, Brazil
| | - Tiago Branquinho Oliveira
- Department of Pharmacy, Federal University of Sergipe (UFS-SE), Av. Marechal Rondon s/n, Jd. Rosa Elze, Sao Cristovao 49100-000, SE, Brazil
| | - Vinicius Barreto da Silva
- Department of Biomedicine and Pharmacy, Pontifical Catholic University of Goiás, 74605-140 Goiania-GO, Brazil
| | - Daniel Fábio Kawano
- Faculty of Pharmaceutical Sciences, University of Campinas, Rua Candido Portinari 200, 13083-871 Campinas- SP, Brazil
| | - Rodrigo Rezende Kitagawa
- Graduate Program in Pharmaceutical Sciences, Health Sciences Center - CCS, Federal University of Espirito Santo - UFES, Marechal Campos Av., 1468, Vitoria 29043-900, ES, Brazil
| |
Collapse
|
15
|
Cinelli MA, Do HT, Miley GP, Silverman RB. Inducible nitric oxide synthase: Regulation, structure, and inhibition. Med Res Rev 2020; 40:158-189. [PMID: 31192483 PMCID: PMC6908786 DOI: 10.1002/med.21599] [Citation(s) in RCA: 464] [Impact Index Per Article: 92.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/14/2019] [Accepted: 05/13/2019] [Indexed: 12/11/2022]
Abstract
A considerable number of human diseases have an inflammatory component, and a key mediator of immune activation and inflammation is inducible nitric oxide synthase (iNOS), which produces nitric oxide (NO) from l-arginine. Overexpressed or dysregulated iNOS has been implicated in numerous pathologies including sepsis, cancer, neurodegeneration, and various types of pain. Extensive knowledge has been accumulated about the roles iNOS plays in different tissues and organs. Additionally, X-ray crystal and cryogenic electron microscopy structures have shed new insights on the structure and regulation of this enzyme. Many potent iNOS inhibitors with high selectivity over related NOS isoforms, neuronal NOS, and endothelial NOS, have been discovered, and these drugs have shown promise in animal models of endotoxemia, inflammatory and neuropathic pain, arthritis, and other disorders. A major issue in iNOS inhibitor development is that promising results in animal studies have not translated to humans; there are no iNOS inhibitors approved for human use. In addition to assay limitations, both the dual modalities of iNOS and NO in disease states (ie, protective vs harmful effects) and the different roles and localizations of NOS isoforms create challenges for therapeutic intervention. This review summarizes the structure, function, and regulation of iNOS, with focus on the development of iNOS inhibitors (historical and recent). A better understanding of iNOS' complex functions is necessary before specific drug candidates can be identified for classical indications such as sepsis, heart failure, and pain; however, newer promising indications for iNOS inhibition, such as depression, neurodegenerative disorders, and epilepsy, have been discovered.
Collapse
Affiliation(s)
- Maris A. Cinelli
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
- Current address: Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824
| | - Ha T. Do
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
- Current address: Mersana Therapeutics, Inc., Cambridge, MA 02139
| | - Galen P. Miley
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
| | - Richard B. Silverman
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
| |
Collapse
|
16
|
Minhas R, Bansal Y, Bansal G. Inducible nitric oxide synthase inhibitors: A comprehensive update. Med Res Rev 2019; 40:823-855. [PMID: 31502681 DOI: 10.1002/med.21636] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 12/27/2022]
Abstract
Inducible nitric oxide synthase (iNOS), which is expressed in response to bacterial/proinflammatory stimuli, generates nitric oxide (NO) that provides cytoprotection. Overexpression of iNOS increases the levels of NO, and this increased NO level is implicated in pathophysiology of complex multifactorial diseases like Parkinson's disease, Alzheimer's disease, multiple sclerosis, rheumatoid arthritis, and inflammatory bowel disease. Selective inhibition of iNOS is an effective approach in treatment of such complex diseases. l-Arginine, being a substrate for iNOS, is the natural lead to develop iNOS inhibitors. More than 200 research reports on development of nitric oxide synthase inhibitors by different research groups across the globe have appeared in literature so far. The first review on iNOS, in 2002, discussed the iNOS inhibitors under two classes that is, amino acid and non-amino acid derivatives. Other review articles discussing specific chemical classes of iNOS inhibitors also appeared during last decade. In the present review, all reports on both natural and synthetic iNOS inhibitors, published 2002 onwards, are studied, classified, and discussed to provide comprehensive information on iNOS inhibitors. The synthetic inhibitors are broadly classified into two categories that is, arginine and non-arginine analogs. The latter are further classified into amidines, five- or six-membered heterocyclics, fused cyclics, steroidal type, and chalcones analogs. Structures of the most/significantly potent compounds from each report are provided to know the functional groups important for incurring iNOS inhibitory activity and selectivity. This review is aimed to provide a comprehensive view to the medicinal chemists for rational designing of novel and potent iNOS inhibitors.
Collapse
Affiliation(s)
- Richa Minhas
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Yogita Bansal
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Gulshan Bansal
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| |
Collapse
|
17
|
Ni D, Lu S, Zhang J. Emerging roles of allosteric modulators in the regulation of protein-protein interactions (PPIs): A new paradigm for PPI drug discovery. Med Res Rev 2019; 39:2314-2342. [PMID: 30957264 DOI: 10.1002/med.21585] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 03/12/2019] [Accepted: 03/24/2019] [Indexed: 12/26/2022]
Abstract
Protein-protein interactions (PPIs) are closely implicated in various types of cellular activities and are thus pivotal to health and disease states. Given their fundamental roles in a wide range of biological processes, the modulation of PPIs has enormous potential in drug discovery. However, owing to the general properties of large, flat, and featureless interfaces of PPIs, previous attempts have demonstrated that the generation of therapeutic agents targeting PPI interfaces is challenging, rendering them almost "undruggable" for decades. To date, rapid progress in chemical and structural biology techniques has promoted the exploitation of allostery as a novel approach in drug discovery. By attaching to allosteric sites that are topologically and spatially distinct from PPI interfaces, allosteric modulators can achieve improved physiochemical properties. Thus, allosteric modulators may represent an alternative strategy to target intractable PPIs and have attracted intense pharmaceutical interest. In this review, we first briefly introduce the characteristics of PPIs and then present different approaches for investigating PPIs, as well as the latest methods for modulating PPIs. Importantly, we comprehensively review the recent progress in the development of allosteric modulators to inhibit or stabilize PPIs. Finally, we conclude with future perspectives on the discovery of allosteric PPI modulators, especially the application of computational methods to aid in allosteric PPI drug discovery.
Collapse
Affiliation(s)
- Duan Ni
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Clinical and Fundamental Research Center, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Shaoyong Lu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Clinical and Fundamental Research Center, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China.,Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Jian Zhang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Clinical and Fundamental Research Center, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China.,Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai, China.,Center for Single-Cell Omics, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Shi JB, Chen LZ, Wang BS, Huang X, Jiao MM, Liu MM, Tang WJ, Liu XH. Novel Pyrazolo[4,3-d]pyrimidine as Potent and Orally Active Inducible Nitric Oxide Synthase (iNOS) Dimerization Inhibitor with Efficacy in Rheumatoid Arthritis Mouse Model. J Med Chem 2019; 62:4013-4031. [DOI: 10.1021/acs.jmedchem.9b00039] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Jing Bo Shi
- School of Pharmacy, Anhui Medical University, Hefei 230032, P. R. China
| | - Liu Zeng Chen
- School of Pharmacy, Anhui Medical University, Hefei 230032, P. R. China
| | - Bao Shi Wang
- School of Pharmacy, Anhui Medical University, Hefei 230032, P. R. China
| | - Xin Huang
- School of Pharmacy, Anhui Medical University, Hefei 230032, P. R. China
| | - Ming Ming Jiao
- School of Pharmacy, Anhui Medical University, Hefei 230032, P. R. China
| | - Ming Ming Liu
- School of Pharmacy, Anhui Medical University, Hefei 230032, P. R. China
| | - Wen Jian Tang
- School of Pharmacy, Anhui Medical University, Hefei 230032, P. R. China
| | - Xin Hua Liu
- School of Pharmacy, Anhui Medical University, Hefei 230032, P. R. China
| |
Collapse
|
19
|
Zhong L, Tran T, Baguley TD, Lee SJ, Henke A, To A, Li S, Yu S, Grieco FA, Roland J, Schultz PG, Eizirik DL, Rogers N, Chartterjee AK, Tremblay MS, Shen W. A novel inhibitor of inducible NOS dimerization protects against cytokine-induced rat beta cell dysfunction. Br J Pharmacol 2018; 175:3470-3485. [PMID: 29888783 PMCID: PMC6086989 DOI: 10.1111/bph.14388] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 05/14/2018] [Accepted: 05/28/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Beta cell apoptosis is a major feature of type 1 diabetes, and pro-inflammatory cytokines are key drivers of the deterioration of beta cell mass through induction of apoptosis. Mitochondrial stress plays a critical role in mediating apoptosis by releasing cytochrome C into the cytoplasm, directly activating caspase-9 and its downstream signalling cascade. We aimed to identify new compounds that protect beta cells from cytokine-induced activation of the intrinsic (mitochondrial) pathway of apoptosis. EXPERIMENTAL APPROACH Diabetogenic media, composed of IL-1β, IFN-γ and high glucose, were used to induce mitochondrial stress in rat insulin-producing INS1E cells, and a high-content image-based screen of small molecule modulators of Casp9 pathway was performed. KEY RESULTS A novel small molecule, ATV399, was identified from a high-content image-based screen for compounds that inhibit cleaved caspase-9 activation and subsequent beta cell apoptosis induced by a combination of IL-1β, IFN-γ and high glucose, which together mimic the pathogenic diabetic milieu. Through medicinal chemistry optimization, potency was markedly improved (6-30 fold), with reduced inhibitory effects on CYP3A4. Improved analogues, such as CAT639, improved beta cell viability and insulin secretion in cytokine-treated rat insulin-producing INS1E cells and primary dispersed islet cells. Mechanistically, CAT639 reduced the production of NO by allosterically inhibiting dimerization of inducible NOS (iNOS) without affecting its mRNA levels. CONCLUSION AND IMPLICATIONS Taken together, these studies demonstrate a successful phenotypic screening campaign resulting in identification of an inhibitor of iNOS dimerization that protects beta cell viability and function through modulation of mitochondrial stress induced by cytokines.
Collapse
Affiliation(s)
- Linlin Zhong
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Tuan Tran
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Tyler D Baguley
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Sang Jun Lee
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Adam Henke
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Andrew To
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Sijia Li
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Shan Yu
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Fabio A Grieco
- ULB Center for Diabetes ResearchUniversite´ Libre de Bruxelles (ULB)Brussels1070Belgium
| | - Jason Roland
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | - Peter G Schultz
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
- Department of ChemistryThe Scripps Research InstituteLa JollaCA92037USA
| | - Decio L Eizirik
- ULB Center for Diabetes ResearchUniversite´ Libre de Bruxelles (ULB)Brussels1070Belgium
| | - Nikki Rogers
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| | | | | | - Weijun Shen
- California Institute for Biomedical Research (Calibr)La JollaCA92037USA
| |
Collapse
|
20
|
Ardisson JS, Gonçalves RDCR, Rodrigues RP, Kitagawa RR. Antitumour, Immunomodulatory activity and in silico studies of naphthopyranones targeting iNOS, a relevant target for the treatment of Helicobacter pylori infection. Biomed Pharmacother 2018; 107:1160-1165. [PMID: 30257329 DOI: 10.1016/j.biopha.2018.08.098] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 01/05/2023] Open
Abstract
The naphthopyranones paepalantine and 5-methoxy-3,4-dehydroxanthomegnin isolated from Paepalanthus sp, showed in previous studies antioxidant, anti-inflammatory, antitumour and antimicrobial potential, such as anti-Helicobacter pylori activity. H. pylori infection is one of the main causes of gastric cancer, causing an excessive inflammatory response through the neutrophils and macrophages infiltration, increasing the release of reactive species and thus inducing the production of pro-inflammatory mediators. In the present study, immunomodulatory activity of naphthopyranones in LPS-stimulated macrophages and cytotoxic action in gastric adenocarcinoma cell lines was evaluated. The potential of interaction of these substances in the iNOS binding site was investigated by molecular docking. Cytotoxic activity in gastric adenocarcinoma cells (AGS) was evaluated by the MTT assay. The results evidenced immunomodulatory potential by inhibiting the pro-inflammatory cytokines and nitric oxide produced by LPS-stimulated macrophages. Cytotoxic activity in AGS cell line was also reported. The results indicated that the studied naphthopyranones are viable alternatives in the treatment and prevention of H. pylori infection as well as the diseases related to this infection, especially gastric cancer.
Collapse
Affiliation(s)
- Juliana Santa Ardisson
- Graduate Program in Pharmaceutical Sciences, Federal University of Espirito Santo - UFES, Avenida Marechal Campos 1468, 29047-105 Vitória, ES, Brazil
| | - Rita de Cássia Ribeiro Gonçalves
- Graduate Program in Pharmaceutical Sciences, Federal University of Espirito Santo - UFES, Avenida Marechal Campos 1468, 29047-105 Vitória, ES, Brazil; Department of Pharmaceutical Sciences, Federal University of Espirito Santo - UFES, Avenida Marechal Campos 1468, 29047-105 Vitória, ES, Brazil
| | - Ricardo Pereira Rodrigues
- Graduate Program in Pharmaceutical Sciences, Federal University of Espirito Santo - UFES, Avenida Marechal Campos 1468, 29047-105 Vitória, ES, Brazil
| | - Rodrigo Rezende Kitagawa
- Graduate Program in Pharmaceutical Sciences, Federal University of Espirito Santo - UFES, Avenida Marechal Campos 1468, 29047-105 Vitória, ES, Brazil; Department of Pharmaceutical Sciences, Federal University of Espirito Santo - UFES, Avenida Marechal Campos 1468, 29047-105 Vitória, ES, Brazil.
| |
Collapse
|
21
|
Bignon E, Allega MF, Lucchetta M, Tiberti M, Papaleo E. Computational Structural Biology of S-nitrosylation of Cancer Targets. Front Oncol 2018; 8:272. [PMID: 30155439 PMCID: PMC6102371 DOI: 10.3389/fonc.2018.00272] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/02/2018] [Indexed: 12/15/2022] Open
Abstract
Nitric oxide (NO) plays an essential role in redox signaling in normal and pathological cellular conditions. In particular, it is well known to react in vivo with cysteines by the so-called S-nitrosylation reaction. S-nitrosylation is a selective and reversible post-translational modification that exerts a myriad of different effects, such as the modulation of protein conformation, activity, stability, and biological interaction networks. We have appreciated, over the last years, the role of S-nitrosylation in normal and disease conditions. In this context, structural and computational studies can help to dissect the complex and multifaceted role of this redox post-translational modification. In this review article, we summarized the current state-of-the-art on the mechanism of S-nitrosylation, along with the structural and computational studies that have helped to unveil its effects and biological roles. We also discussed the need to move new steps forward especially in the direction of employing computational structural biology to address the molecular and atomistic details of S-nitrosylation. Indeed, this redox modification has been so far an underappreciated redox post-translational modification by the computational biochemistry community. In our review, we primarily focus on S-nitrosylated proteins that are attractive cancer targets due to the emerging relevance of this redox modification in a cancer setting.
Collapse
Affiliation(s)
- Emmanuelle Bignon
- Computational Biology Laboratory Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Maria Francesca Allega
- Computational Biology Laboratory Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Marta Lucchetta
- Computational Biology Laboratory Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Matteo Tiberti
- Computational Biology Laboratory Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Elena Papaleo
- Computational Biology Laboratory Danish Cancer Society Research Center, Copenhagen, Denmark.,Translational Disease Systems Biology, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
A small-molecule inhibitor of SOD1-Derlin-1 interaction ameliorates pathology in an ALS mouse model. Nat Commun 2018; 9:2668. [PMID: 29991716 PMCID: PMC6039432 DOI: 10.1038/s41467-018-05127-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder. Despite its severity, there are no effective treatments because of the complexity of its pathogenesis. As one of the underlying mechanisms of Cu, Zn superoxide dismutase (SOD1) gene mutation-induced ALS, SOD1 mutants (SOD1mut) commonly interact with an endoplasmic reticulum-resident membrane protein Derlin-1, triggering motoneuron death. However, the importance of SOD1-Derlin-1 interaction in in vitro human model and in vivo mouse model remains to be elucidated. Here, we identify small-molecular-weight compounds that inhibit the SOD1-Derlin-1 interaction by screening approximately 160,000 compounds. The inhibitor prevents 122 types of SOD1mut from interacting with Derlin-1, and significantly ameliorates the ALS pathology both in motoneurons derived from patient induced pluripotent stem cells and in model mice. Our data suggest that the SOD1-Derlin-1 interaction contributes to the pathogenesis of ALS and is a promising drug target for ALS treatment. Amyotrophic lateral sclerosis (ALS) is a neurological disease that leads to loss of voluntary muscle movement. Here, the authors screen for molecules that disrupt interaction between SOD1, a protein linked to ALS, and Derlin-1, and find an inhibitor that reduces pathology in an ALS mouse model.
Collapse
|
23
|
Valeur E, Jimonet P. New Modalities, Technologies, and Partnerships in Probe and Lead Generation: Enabling a Mode-of-Action Centric Paradigm. J Med Chem 2018; 61:9004-9029. [DOI: 10.1021/acs.jmedchem.8b00378] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Eric Valeur
- Medicinal Chemistry, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal 431 83, Sweden
| | - Patrick Jimonet
- External Innovation Drug Discovery, Global Business Development & Licensing, Sanofi, 13 quai Jules Guesde, 94400 Vitry-sur-Seine, France
| |
Collapse
|
24
|
Llano M, Peña-Hernandez MA. Defining Pharmacological Targets by Analysis of Virus-Host Protein Interactions. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 111:223-242. [PMID: 29459033 PMCID: PMC6322211 DOI: 10.1016/bs.apcsb.2017.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Viruses are obligate parasites that depend on cellular factors for replication. Pharmacological inhibition of essential viral proteins, mostly enzymes, is an effective therapeutic alternative in the absence of effective vaccines. However, this strategy commonly encounters drug resistance mechanisms that allow these pathogens to evade control. Due to the dependency on host factors for viral replication, pharmacological disruption of the host-pathogen protein-protein interactions (PPIs) is an important therapeutic alternative to block viral replication. In this review we discuss salient aspects of PPIs implicated in viral replication and advances in the development of small molecules that inhibit viral replication through antagonism of these interactions.
Collapse
Affiliation(s)
- Manuel Llano
- University of Texas at El Paso, El Paso, TX, United States.
| | - Mario A Peña-Hernandez
- University of Texas at El Paso, El Paso, TX, United States; Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolas de los Garza, Mexico
| |
Collapse
|
25
|
|
26
|
Rashidi B, Hoseini Z, Sahebkar A, Mirzaei H. Anti-Atherosclerotic Effects of Vitamins D and E in Suppression of Atherogenesis. J Cell Physiol 2017; 232:2968-2976. [DOI: 10.1002/jcp.25738] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 12/13/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Bahman Rashidi
- Department of Anatomical Sciences and Molecular Biology; School of Medicine; Isfahan University of Medical Sciences; Isfahan Iran
| | - Zahra Hoseini
- Student Research Center; School of Medicine; Isfahan University of Medical Sciences; Isfahan Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| | - Hamed Mirzaei
- Department of Medical Biotechnology; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
27
|
Poulos TL, Li H. Nitric oxide synthase and structure-based inhibitor design. Nitric Oxide 2016; 63:68-77. [PMID: 27890696 DOI: 10.1016/j.niox.2016.11.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 11/09/2016] [Accepted: 11/21/2016] [Indexed: 11/24/2022]
Abstract
Once it was discovered that the enzyme nitric oxide synthase (NOS) is responsible for the biosynthesis of NO, NOS became a drug target. Particularly important is the over production of NO by neuronal NOS (nNOS) in various neurodegenerative disorders. After the various NOS isoforms were identified, inhibitor development proceeded rapidly. It soon became evident, however, that isoform selectivity presents a major challenge. All 3 human NOS isoforms, nNOS, eNOS (endothelial NOS), and iNOS (inducible NOS) have nearly identical active site structures thus making selective inhibitor design especially difficult. Of particular importance is the avoidance of inhibiting eNOS owing to its vital role in the cardiovascular system. This review summarizes some of the history of NOS inhibitor development and more recent advances in developing isoform selective inhibitors using primarily structure-based approaches.
Collapse
Affiliation(s)
- Thomas L Poulos
- Departments of Molecular Biology & Biochemistry, Pharmaceutical Sciences, and Chemistry, University of California, Irvine, Irvine, CA 92697-3900, USA.
| | - Huiying Li
- Departments of Molecular Biology & Biochemistry, Pharmaceutical Sciences, and Chemistry, University of California, Irvine, Irvine, CA 92697-3900, USA
| |
Collapse
|
28
|
Seetoh WG, Abell C. Disrupting the Constitutive, Homodimeric Protein-Protein Interface in CK2β Using a Biophysical Fragment-Based Approach. J Am Chem Soc 2016; 138:14303-14311. [PMID: 27726344 PMCID: PMC5257173 DOI: 10.1021/jacs.6b07440] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
![]()
Identifying small molecules that
induce the disruption of constitutive
protein–protein interfaces is a challenging objective. Here,
a targeted biophysical screening cascade was employed to specifically
identify small molecules that could disrupt the constitutive, homodimeric
protein–protein interface within CK2β. This approach
could potentially be applied to achieve subunit disassembly of other
homo-oligomeric proteins as a means of modulating protein function.
Collapse
Affiliation(s)
- Wei-Guang Seetoh
- Department of Chemistry, University of Cambridge , Lensfield Road, Cambridge, CB2 1EW, United Kingdom
| | - Chris Abell
- Department of Chemistry, University of Cambridge , Lensfield Road, Cambridge, CB2 1EW, United Kingdom
| |
Collapse
|
29
|
Abstract
Modulation of protein-protein interactions (PPIs) is becoming increasingly important in drug discovery and chemical biology. While a few years ago this 'target class' was deemed to be largely undruggable an impressing number of publications and success stories now show that targeting PPIs with small, drug-like molecules indeed is a feasible approach. Here, we summarize the current state of small-molecule inhibition and stabilization of PPIs and review the active molecules from a structural and medicinal chemistry angle, especially focusing on the key examples of iNOS, LFA-1 and 14-3-3.
Collapse
|
30
|
Small Molecule Targeting of Protein-Protein Interactions through Allosteric Modulation of Dynamics. Molecules 2015; 20:16435-45. [PMID: 26378508 PMCID: PMC6332300 DOI: 10.3390/molecules200916435] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/06/2015] [Accepted: 09/08/2015] [Indexed: 11/16/2022] Open
Abstract
The protein–protein interaction (PPI) target class is particularly challenging, but offers potential for “first in class” therapies. Most known PPI small molecules are orthosteric inhibitors but many PPI sites may be fundamentally intractable to this approach. One potential alternative is to consider more attractive, remote small molecule pockets; however, on the whole, allostery is poorly understood and difficult to discover and develop. Here we review the literature in order to understand the basis for allostery, especially as it can apply to PPIs. We suggest that the upfront generation of sophisticated and experimentally validated dynamic models of target proteins can aid in target choice and strategy for allosteric intervention to produce the required functional effect.
Collapse
|
31
|
Oversø Hansen P, Kringelholt S, Simonsen U, Bek T. Hypoxia-induced relaxation of porcine retinal arterioles in vitro depends on inducible NO synthase and EP4 receptor stimulation in the perivascular retina. Acta Ophthalmol 2015; 93:457-463. [PMID: 25619924 DOI: 10.1111/aos.12669] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 12/24/2014] [Indexed: 01/26/2023]
Abstract
PURPOSE Hypoxia-induced relaxation of porcine retinal arterioles has been shown to be reduced during inhibition of prostaglandin synthesis and nitric oxide synthase (NOS). The purpose of this study was to identity the specific prostaglandin receptor(s) and source(s) of NO mediating this effect. METHODS Porcine retinal arterioles with preserved perivascular retinal tissue were mounted in a myograph and were exposed to hypoxia in the presence of one of the following: the general NO synthase inhibitor L-NAME, the selective iNOS inhibitor 1400W, the selective nNOS inhibitor 7-nitroindazole, the general cyclooxygenase (COX) inhibitor ibuprofen or an antagonist to the FP- (AL 8810), DP- (BWA868C), EP1 - (SC-19220), EP2 - (PF-044189) or EP4 receptors (GW627368X). The experiments were repeated after removal of the perivascular retinal tissue. RESULTS Hypoxia induced relaxation of retinal arterioles with preserved perivascular retinal tissue. This relaxation was significantly reduced in the presence of L-NAME, 1400W, ibuprofen and the EP4 receptor antagonist GW627368X. The simultaneous addition of L-NAME or 1400W in combination with ibuprofen, but not GW627368X, reduced hypoxia-induced vasorelaxation additively as compared to the effect of the compounds individually. CONCLUSION Hypoxia-induced vasorelaxation of porcine retinal arterioles is mediated by inducible NOS and stimulation of EP4 receptors acting through separate pathways, but mechanisms unrelated to the studied prostaglandin receptors and NOS products are also involved.
Collapse
Affiliation(s)
| | - Sidse Kringelholt
- Department of Ophthalmology; Aarhus University Hospital; Aarhus Denmark
| | - Ulf Simonsen
- Department of Biomedicine; University of Aarhus; Aarhus Denmark
| | - Toke Bek
- Department of Ophthalmology; Aarhus University Hospital; Aarhus Denmark
| |
Collapse
|
32
|
Hamdi A, Lesnard A, Suzanne P, Robert T, Miteva MA, Pellerano M, Didier B, Ficko-Blean E, Lobstein A, Hibert M, Rault S, Morris MC, Colas P. Tampering with Cell Division by Using Small-Molecule Inhibitors of CDK-CKS Protein Interactions. Chembiochem 2015; 16:432-9. [DOI: 10.1002/cbic.201402579] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Indexed: 11/07/2022]
|
33
|
Mukherjee P, Li H, Sevrioukova I, Chreifi G, Martásek P, Roman LJ, Poulos TL, Silverman RB. Novel 2,4-disubstituted pyrimidines as potent, selective, and cell-permeable inhibitors of neuronal nitric oxide synthase. J Med Chem 2014; 58:1067-88. [PMID: 25489882 PMCID: PMC4329833 DOI: 10.1021/jm501719e] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Selective inhibition of neuronal nitric oxide synthase (nNOS) is an important therapeutic approach to target neurodegenerative disorders. However, the majority of the nNOS inhibitors developed are arginine mimetics and, therefore, suffer from poor bioavailability. We designed a novel strategy to combine a more pharmacokinetically favorable 2-imidazolylpyrimidine head with promising structural components from previous inhibitors. In conjunction with extensive structure-activity studies, several highly potent and selective inhibitors of nNOS were discovered. X-ray crystallographic analysis reveals that these type II inhibitors utilize the same hydrophobic pocket to gain strong inhibitory potency (13), as well as high isoform selectivity. Interestingly, select compounds from this series (9) showed good permeability and low efflux in a Caco-2 assay, suggesting potential oral bioavailability, and exhibited minimal off-target binding to 50 central nervous system receptors. Furthermore, even with heme-coordinating groups in the molecule, modifying other pharmacophoric fragments minimized undesirable inhibition of cytochrome P450s from human liver microsomes.
Collapse
Affiliation(s)
- Paramita Mukherjee
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Northwestern University , Evanston, Illinois 60208-3113, United States
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Mukherjee P, Cinelli MA, Kang S, Silverman RB. Development of nitric oxide synthase inhibitors for neurodegeneration and neuropathic pain. Chem Soc Rev 2014; 43:6814-38. [PMID: 24549364 PMCID: PMC4138306 DOI: 10.1039/c3cs60467e] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Nitric oxide (NO) is an important signaling molecule in the human body, playing a crucial role in cell and neuronal communication, regulation of blood pressure, and in immune activation. However, overproduction of NO by the neuronal isoform of nitric oxide synthase (nNOS) is one of the fundamental causes underlying neurodegenerative disorders and neuropathic pain. Therefore, developing small molecules for selective inhibition of nNOS over related isoforms (eNOS and iNOS) is therapeutically desirable. The aims of this review focus on the regulation and dysregulation of NO signaling, the role of NO in neurodegeneration and pain, the structure and mechanism of nNOS, and the use of this information to design selective inhibitors of this enzyme. Structure-based drug design, the bioavailability and pharmacokinetics of these inhibitors, and extensive target validation through animal studies are addressed.
Collapse
Affiliation(s)
- Paramita Mukherjee
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113, USA.
| | | | | | | |
Collapse
|
35
|
Pei J, Yin N, Ma X, Lai L. Systems Biology Brings New Dimensions for Structure-Based Drug Design. J Am Chem Soc 2014; 136:11556-65. [DOI: 10.1021/ja504810z] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jianfeng Pei
- Center
for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Ning Yin
- Center
for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Xiaomin Ma
- Center
for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Luhua Lai
- Center
for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Beijing
National Laboratory for Molecular Science, State Key Laboratory for
Structural Chemistry of Unstable and Stable Species, College of Chemistry
and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua
Center for Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
36
|
Mukherjee M, Jing-Song F, Ramachandran S, Guy GR, Sivaraman J. Dimeric switch of Hakai-truncated monomers during substrate recognition: insights from solution studies and NMR structure. J Biol Chem 2014; 289:25611-23. [PMID: 25074933 DOI: 10.1074/jbc.m114.592840] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hakai, an E3 ubiquitin ligase, disrupts cell-cell contacts in epithelial cells and is up-regulated in human colon and gastric adenocarcinomas. Hakai acts through its phosphotyrosine-binding (HYB) domain, which bears a dimeric fold that recognizes the phosphotyrosine motifs of E-cadherin, cortactin, DOK1, and other Src substrates. Unlike the monomeric nature of the SH2 and phosphotyrosine-binding domains, the architecture of the HYB domain consists of an atypical, zinc-coordinated tight homodimer. Here, we report a C-terminal truncation mutant of the HYB domain (HYB(ΔC)), comprising amino acids 106-194, which exists as a monomer in solution. The NMR structure revealed that this deletion mutant undergoes a dramatic structural change caused by a rearrangement of the atypical zinc-coordinated unit in the C terminus of the HYB domain to a C2H2-like zinc finger in HYB(ΔC). Moreover, using isothermal titration calorimetry, we show that dimerization of HYB(ΔC) can be induced using a phosphotyrosine substrate peptide. This ligand-induced dimerization of HYB(ΔC) is further validated using analytical ultracentrifugation, size-exclusion chromatography, NMR relaxation studies, dynamic light scattering, and circular dichroism experiments. Overall, these observations suggest that the dimeric architecture of the HYB domain is essential for the phosphotyrosine-binding property of Hakai.
Collapse
Affiliation(s)
- Manjeet Mukherjee
- From the Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543 and
| | - Fan Jing-Song
- From the Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543 and
| | - Sarath Ramachandran
- From the Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543 and
| | - Graeme R Guy
- the Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673
| | - J Sivaraman
- From the Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543 and
| |
Collapse
|
37
|
Nero TL, Morton CJ, Holien JK, Wielens J, Parker MW. Oncogenic protein interfaces: small molecules, big challenges. Nat Rev Cancer 2014; 14:248-62. [PMID: 24622521 DOI: 10.1038/nrc3690] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Historically, targeting protein-protein interactions with small molecules was not thought possible because the corresponding interfaces were considered mostly flat and featureless and therefore 'undruggable'. Instead, such interactions were targeted with larger molecules, such as peptides and antibodies. However, the past decade has seen encouraging breakthroughs through the refinement of existing techniques and the development of new ones, together with the identification and exploitation of unexpected aspects of protein-protein interaction surfaces. In this Review, we describe some of the latest techniques to discover modulators of protein-protein interactions and how current drug discovery approaches have been adapted to successfully target these interfaces.
Collapse
Affiliation(s)
- Tracy L Nero
- Australian Cancer Research Foundation Rational Drug Discovery Centre and Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Victoria 3065, Australia
| | - Craig J Morton
- Australian Cancer Research Foundation Rational Drug Discovery Centre and Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Victoria 3065, Australia
| | - Jessica K Holien
- Australian Cancer Research Foundation Rational Drug Discovery Centre and Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Victoria 3065, Australia
| | - Jerome Wielens
- 1] Australian Cancer Research Foundation Rational Drug Discovery Centre and Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Victoria 3065, Australia. [2] Department of Medicine, University of Melbourne, 41 Victoria Parade, Fitzroy, Victoria 3065, Australia
| | - Michael W Parker
- 1] Australian Cancer Research Foundation Rational Drug Discovery Centre and Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Victoria 3065, Australia. [2] Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
38
|
Zhong HJ, Liu LJ, Chong CM, Lu L, Wang M, Chan DSH, Chan PWH, Lee SMY, Ma DL, Leung CH. Discovery of a natural product-like iNOS inhibitor by molecular docking with potential neuroprotective effects in vivo. PLoS One 2014; 9:e92905. [PMID: 24690920 PMCID: PMC3972188 DOI: 10.1371/journal.pone.0092905] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 02/27/2014] [Indexed: 12/11/2022] Open
Abstract
In this study, we applied structure-based virtual screening techniques to identify natural product or natural product-like inhibitors of iNOS. The iNOS inhibitory activity of the hit compounds was characterized using cellular assays and an in vivo zebrafish larvae model. The natural product-like compound 1 inhibited NO production in LPS-stimulated Raw264.7 macrophages, without exerting cytotoxic effects on the cells. Significantly, compound 1 was able to reverse MPTP-induced locomotion deficiency and neurotoxicity in an in vivo zebrafish larval model. Hence, compound 1 could be considered as a scaffold for the further development of iNOS inhibitors for potential anti-inflammatory or anti-neurodegenerative applications.
Collapse
Affiliation(s)
- Hai-Jing Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Li-Juan Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Cheong-Meng Chong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Lihua Lu
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Modi Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Daniel Shiu-Hin Chan
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Philip Wai Hong Chan
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore, Singapore
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
39
|
Doi T, Yoshida M, Ohsawa K, Shin-ya K, Takagi M, Uekusa Y, Yamaguchi T, Kato K, Hirokawa T, Natsume T. Total synthesis and characterization of thielocin B1 as a protein–protein interaction inhibitor of PAC3 homodimer. Chem Sci 2014. [DOI: 10.1039/c3sc53237b] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We have characterized the inhibition of the protein–protein interaction of the homodimer of proteasome assembling chaperone (PAC) 3 with thielocin B1.
Collapse
Affiliation(s)
- Takayuki Doi
- Graduate School of Pharmaceutical Sciences
- Tohoku University
- Sendai 980-8578, Japan
| | - Masahito Yoshida
- Graduate School of Pharmaceutical Sciences
- Tohoku University
- Sendai 980-8578, Japan
| | - Kosuke Ohsawa
- Graduate School of Pharmaceutical Sciences
- Tohoku University
- Sendai 980-8578, Japan
| | - Kazuo Shin-ya
- National Institute of Advanced Industrial Science and Technology (AIST)
- Tokyo 135-0064, Japan
| | - Motoki Takagi
- Biomedicinal Information Research Center (BIRC)
- Japan Biological Informatics Consortium (JBIC)
- Tokyo 135-0064, Japan
| | - Yoshinori Uekusa
- Okazaki Institute for Integrative Bioscience and Institute for Molecular Science
- National Institutes of Natural Sciences
- Okazaki, Japan
- Graduate School of Pharmaceutical Sciences
- Nagoya City University
| | - Takumi Yamaguchi
- Okazaki Institute for Integrative Bioscience and Institute for Molecular Science
- National Institutes of Natural Sciences
- Okazaki, Japan
- Graduate School of Pharmaceutical Sciences
- Nagoya City University
| | - Koichi Kato
- Okazaki Institute for Integrative Bioscience and Institute for Molecular Science
- National Institutes of Natural Sciences
- Okazaki, Japan
- Graduate School of Pharmaceutical Sciences
- Nagoya City University
| | - Takatsugu Hirokawa
- Computational Biology Research Center (CBRC)
- National Institute of Advanced Industrial Science and Technology (AIST)
- Tokyo 135-0064, Japan
| | - Tohru Natsume
- National Institute of Advanced Industrial Science and Technology (AIST)
- Tokyo 135-0064, Japan
| |
Collapse
|
40
|
Ohkanda J. Module Assembly for Designing Multivalent Mid-Sized Inhibitors of Protein-Protein Interactions. CHEM REC 2013; 13:561-75. [DOI: 10.1002/tcr.201300026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Junko Ohkanda
- Institute for Chemical Research; Kyoto University; Gokasho, Uji Kyoto 611-0011 Japan
| |
Collapse
|
41
|
Mignani S, El Kazzouli S, Bousmina MM, Majoral JP. Dendrimer Space Exploration: An Assessment of Dendrimers/Dendritic Scaffolding as Inhibitors of Protein–Protein Interactions, a Potential New Area of Pharmaceutical Development. Chem Rev 2013; 114:1327-42. [DOI: 10.1021/cr400362r] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Serge Mignani
- Laboratoire de Chimie et de
Biochimie Pharmacologiques
et Toxicologiques, CNRS UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints Pères, 75006 Paris, France
| | - Saïd El Kazzouli
- Euro-Mediterranean University of Fez, Fès-Shore, Route de Sidi harazem, Fès, Morocco
| | - Mosto M. Bousmina
- Euro-Mediterranean University of Fez, Fès-Shore, Route de Sidi harazem, Fès, Morocco
- Hassan II Academy of Science and Technology, Avenue Mohammed
VI, 10222 Rabat, Morocco
| | - Jean-Pierre Majoral
- Laboratoire
de Chimie de Coordination, Centre National de la Recherche Scientifique, 205 route de Narbonne, 31077 Toulouse Cedex 4, France
| |
Collapse
|
42
|
Nagpal L, Haque MM, Saha A, Mukherjee N, Ghosh A, Ranu BC, Stuehr DJ, Panda K. Mechanism of inducible nitric-oxide synthase dimerization inhibition by novel pyrimidine imidazoles. J Biol Chem 2013; 288:19685-97. [PMID: 23696643 PMCID: PMC3707674 DOI: 10.1074/jbc.m112.446542] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 05/13/2013] [Indexed: 01/19/2023] Open
Abstract
Overproduction of nitric oxide (NO) by inducible nitric-oxide synthase (iNOS) has been etiologically linked to several inflammatory, immunological, and neurodegenerative diseases. As dimerization of NOS is required for its activity, several dimerization inhibitors, including pyrimidine imidazoles, are being evaluated for therapeutic inhibition of iNOS. However, the precise mechanism of their action is still unclear. Here, we examined the mechanism of iNOS inhibition by a pyrimidine imidazole core compound and its derivative (PID), having low cellular toxicity and high affinity for iNOS, using rapid stopped-flow kinetic, gel filtration, and spectrophotometric analysis. PID bound to iNOS heme to generate an irreversible PID-iNOS monomer complex that could not be converted to active dimers by tetrahydrobiopterin (H4B) and l-arginine (Arg). We utilized the iNOS oxygenase domain (iNOSoxy) and two monomeric mutants whose dimerization could be induced (K82AiNOSoxy) or not induced (D92AiNOSoxy) with H4B to elucidate the kinetics of PID binding to the iNOS monomer and dimer. We observed that the apparent PID affinity for the monomer was 11 times higher than the dimer. PID binding rate was also sensitive to H4B and Arg site occupancy. PID could also interact with nascent iNOS monomers in iNOS-synthesizing RAW cells, to prevent their post-translational dimerization, and it also caused irreversible monomerization of active iNOS dimers thereby accomplishing complete physiological inhibition of iNOS. Thus, our study establishes PID as a versatile iNOS inhibitor and therefore a potential in vivo tool for examining the causal role of iNOS in diseases associated with its overexpression as well as therapeutic control of such diseases.
Collapse
Affiliation(s)
- Latika Nagpal
- From the Department of Biotechnology and Guha Center for Genetic Engineering and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700019, India
| | - Mohammad M. Haque
- the Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, and
| | - Amit Saha
- the Department of Organic Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Nirmalya Mukherjee
- the Department of Organic Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Arnab Ghosh
- the Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, and
| | - Brindaban C. Ranu
- the Department of Organic Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Dennis J. Stuehr
- the Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, and
| | - Koustubh Panda
- From the Department of Biotechnology and Guha Center for Genetic Engineering and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700019, India
| |
Collapse
|
43
|
Immekus F, Barandun LJ, Betz M, Debaene F, Petiot S, Sanglier-Cianferani S, Reuter K, Diederich F, Klebe G. Launching spiking ligands into a protein-protein interface: a promising strategy to destabilize and break interface formation in a tRNA modifying enzyme. ACS Chem Biol 2013; 8:1163-78. [PMID: 23534552 DOI: 10.1021/cb400020b] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Apart from competitive active-site inhibition of protein function, perturbance of protein-protein interactions by small molecules in oligodomain enzymes opens new perspectives for innovative therapeutics. tRNA-guanine transglycosylase (TGT), a potential target to treat shigellosis, is active only as the homodimer. Consequently, disruption of the dimer interface by small molecules provides a novel inhibition mode. A special feature of this enzyme is the short distance between active site and rim of the dimer interface. This suggests design of expanded active-site inhibitors decorated with rigid, needle-type substituents to spike into potential hot spots of the interaction interface. Ligands with attached ethinyl-type substituents have been synthesized and characterized by Kd measurements, crystallography, noncovalent mass spectrometry, and computer simulations. In contrast to previously determined crystal structures with nonextended active-site inhibitors, a well-defined loop-helix motif, involved in several contacts across the dimer interface, falls apart and suggests enhanced flexibility once the spiking ligands are bound. Mass spectrometry indicates significant destabilization but not full disruption of the complexed TGT homodimer in solution. As directed interactions of the loop-helix motif obviously do not determine dimer stability, a structurally conserved hydrophobic patch composed of several aromatic amino acids is suggested as interaction hot spot. The residues of this patch reside on a structurally highly conserved helix-turn-helix motif, which remains unaffected by the bound spiking ligands. Nevertheless, it is shielded from solvent access by the loop-helix motif that becomes perturbed upon binding of the spiking ligands, which serves as a possible explanation for reduced interface stability.
Collapse
Affiliation(s)
- Florian Immekus
- Institut für Pharmazeutische
Chemie, Philipps-Universität Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - Luzi Jakob Barandun
- Laboratorium für Organische
Chemie, ETH Zurich Hönggerberg, HCI, Wolfgang-Pauli-Strasse 10, 8093 Zurich, Switzerland
| | - Michael Betz
- Institut für Pharmazeutische
Chemie, Philipps-Universität Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - François Debaene
- Laboratoire de Spectrométrie de Masse Bio-Organique de Strasbourg, 25, rue Becquerel,
67087 Strasbourg Cedex 2, France
| | - Stéphanie Petiot
- Laboratoire de Spectrométrie de Masse Bio-Organique de Strasbourg, 25, rue Becquerel,
67087 Strasbourg Cedex 2, France
| | - Sarah Sanglier-Cianferani
- Laboratoire de Spectrométrie de Masse Bio-Organique de Strasbourg, 25, rue Becquerel,
67087 Strasbourg Cedex 2, France
| | - Klaus Reuter
- Institut für Pharmazeutische
Chemie, Philipps-Universität Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - François Diederich
- Laboratorium für Organische
Chemie, ETH Zurich Hönggerberg, HCI, Wolfgang-Pauli-Strasse 10, 8093 Zurich, Switzerland
| | - Gerhard Klebe
- Institut für Pharmazeutische
Chemie, Philipps-Universität Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| |
Collapse
|
44
|
Leach AG, Olsson LL, Warner DJ. A monomeric form of iNOS can rationalise observed SAR for inhibitors of dimerisation: quantum mechanics and docking compared. MEDCHEMCOMM 2013. [DOI: 10.1039/c2md20159c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Many interaction types bind iNOS and inhibitors of its dimerization: docking methods reproduce geometries and quantum mechanics lipophilicity ligand efficiencies.
Collapse
|
45
|
A combination of 3D-QSAR, molecular docking and molecular dynamics simulation studies of benzimidazole-quinolinone derivatives as iNOS inhibitors. Int J Mol Sci 2012; 13:11210-11227. [PMID: 23109848 PMCID: PMC3472740 DOI: 10.3390/ijms130911210] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 07/16/2012] [Accepted: 07/30/2012] [Indexed: 12/14/2022] Open
Abstract
Inducible Nitric Oxide Synthase (iNOS) has been involved in a variety of diseases, and thus it is interesting to discover and optimize new iNOS inhibitors. In previous studies, a series of benzimidazole-quinolinone derivatives with high inhibitory activity against human iNOS were discovered. In this work, three-dimensional quantitative structure-activity relationships (3D-QSAR), molecular docking and molecular dynamics (MD) simulation approaches were applied to investigate the functionalities of active molecular interaction between these active ligands and iNOS. A QSAR model with R2 of 0.9356, Q2 of 0.8373 and Pearson-R value of 0.9406 was constructed, which presents a good predictive ability in both internal and external validation. Furthermore, a combined analysis incorporating the obtained model and the MD results indicates: (1) compounds with the proper-size hydrophobic substituents at position 3 in ring-C (R3 substituent), hydrophilic substituents near the X6 of ring-D and hydrophilic or H-bond acceptor groups at position 2 in ring-B show enhanced biological activities; (2) Met368, Trp366, Gly365, Tyr367, Phe363, Pro344, Gln257, Val346, Asn364, Met349, Thr370, Glu371 and Tyr485 are key amino acids in the active pocket, and activities of iNOS inhibitors are consistent with their capability to alter the position of these important residues, especially Glu371 and Thr370. The results provide a set of useful guidelines for the rational design of novel iNOS inhibitors.
Collapse
|
46
|
Thompson AD, Dugan A, Gestwicki JE, Mapp AK. Fine-tuning multiprotein complexes using small molecules. ACS Chem Biol 2012; 7:1311-20. [PMID: 22725693 DOI: 10.1021/cb300255p] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Multiprotein complexes such as the transcriptional machinery, signaling hubs, and protein folding machines are typically composed of at least one enzyme combined with multiple non-enzymes. Often the components of these complexes are incorporated in a combinatorial manner, in which the ultimate composition of the system helps dictate the type, location, or duration of cellular activities. Although drugs and chemical probes have traditionally targeted the enzyme components, emerging strategies call for controlling the function of protein complexes by modulation of protein-protein interactions (PPIs). However, the challenges of targeting PPIs have been well documented, and the diversity of PPIs makes a "one-size-fits-all" solution highly unlikely. These hurdles are particularly daunting for PPIs that encompass large buried surface areas and those with weak affinities. In this Review, we discuss lessons from natural systems, in which allostery and other mechanisms are used to overcome the challenge of regulating the most difficult PPIs. These systems may provide a blueprint for identifying small molecules that target challenging PPIs and affecting molecular decision-making within multiprotein systems.
Collapse
Affiliation(s)
- Andrea D. Thompson
- Program
in Chemical Biology, ‡Departments of Pathology and Biological Chemistry and the Life Sciences
Institute, and §Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Amanda Dugan
- Program
in Chemical Biology, ‡Departments of Pathology and Biological Chemistry and the Life Sciences
Institute, and §Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jason E. Gestwicki
- Program
in Chemical Biology, ‡Departments of Pathology and Biological Chemistry and the Life Sciences
Institute, and §Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Anna K. Mapp
- Program
in Chemical Biology, ‡Departments of Pathology and Biological Chemistry and the Life Sciences
Institute, and §Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
47
|
Features of protein-protein interactions that translate into potent inhibitors: topology, surface area and affinity. Expert Rev Mol Med 2012; 14:e16. [PMID: 22831787 DOI: 10.1017/erm.2012.10] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Protein-protein interactions (PPIs) control the assembly of multi-protein complexes and, thus, these contacts have enormous potential as drug targets. However, the field has produced a mix of both exciting success stories and frustrating challenges. Here, we review known examples and explore how the physical features of a PPI, such as its affinity, hotspots, off-rates, buried surface area and topology, might influence the chances of success in finding inhibitors. This analysis suggests that concise, tight binding PPIs are most amenable to inhibition. However, it is also clear that emerging technical methods are expanding the repertoire of 'druggable' protein contacts and increasing the odds against difficult targets. In particular, natural product-like compound libraries, high throughput screens specifically designed for PPIs and approaches that favour discovery of allosteric inhibitors appear to be attractive routes. The first group of PPI inhibitors has entered clinical trials, further motivating the need to understand the challenges and opportunities in pursuing these types of targets.
Collapse
|
48
|
Suaifan GARY, Shehadehh M, Al-Ijel H, Taha MO. Extensive ligand-based modeling and in silico screening reveal nanomolar inducible nitric oxide synthase (iNOS) inhibitors. J Mol Graph Model 2012; 37:1-26. [PMID: 22609742 DOI: 10.1016/j.jmgm.2012.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Revised: 02/20/2012] [Accepted: 04/02/2012] [Indexed: 01/21/2023]
Abstract
Inducible nitric oxide synthase (iNOS) has been implicated in a variety of diseases prompting several attempts to discover and optimize new iNOS inhibitors. Accordingly, we explored the pharmacophoric space of 143 iNOS inhibitors. Subsequently, genetic algorithm and multiple linear regression analysis were employed to select an optimal combination of pharmacophoric models and 2D physicochemical descriptors to produce self-consistent quantitative structure-activity relationship (QSAR) of optimal predictive potential (correlation coefficient r₁₁₅=0.83, F=23.92, r²(LOO)=0.61, r²(PRESS) against 28 external test inhibitors=0.51). Two orthogonal pharmacophores emerged in the QSAR equation suggesting the existence of at least two binding modes accessible to ligands within iNOS binding pocket. The pharmacophores were validated by comparison with crystallographic complexes of active iNOS inhibitors and receiver operating characteristic (ROC) curves analysis. We employed the pharmacophoric models and associated QSAR equation to screen the national cancer institute list of compounds (NCI). Three low nanomolar inhibitors were identified. The most potent hit exhibited irreversible inhibition of iNOS with IC₅₀ value of 1.4 nM.
Collapse
Affiliation(s)
- Ghadeer A R Y Suaifan
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, Jordan
| | | | | | | |
Collapse
|
49
|
Lee KS, Lee DK, Jeoung D, Lee H, Choe J, Ha KS, Won MH, Kwon YG, Kim YM. Differential effects of substrate-analogue inhibitors on nitric oxide synthase dimerization. Biochem Biophys Res Commun 2012; 418:49-55. [PMID: 22240022 DOI: 10.1016/j.bbrc.2011.12.123] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 12/26/2011] [Indexed: 10/14/2022]
Abstract
Nitric oxide synthase (NOS) isoforms are hemoenzymes that are only active as homodimers. We have examined the effect of the substrate-analogue inhibitors, N(G)-monomethyl-L-arginine (L-NMA), N(G)-nitro-L-arginine (L-NNA), N(G)-nitro-L-arginine methyl ester (L-NAME), N(5)-(1-iminoethyl)-L-ornithine (L-NIO), and N(6)-(1-iminoethyl)-L-lysine (L-NIL), the guanidine-containing inhibitor aminoguanidine (AG), and the amidine moiety-containing iNOS-specific inhibitor 1400W, on the formation of NOS dimer. Of these inhibitors, L-NMA effectively not only inhibited iNOS dimerization, but also destabilized its dimeric form in RAW264.7 cells stimulated with lipopolysaccharide plus interferon-γ, but not eNOS dimerization in endothelial cells. Importantly, this inhibition was highly correlated with NO production. These inhibitory effects were significantly reversed by addition of L-arginine. However, L-NNA, L-NAME, and AG in part or significantly increased dimerization of iNOS and eNOS in intact cells, and the other inhibitors assessed did not alter dimerization of iNOS and eNOS. These data taken together suggest that substituted groups of an arginine guanidino moiety play an important role in NOS dimerization as well as its catalytic activity. Our results indicate that l-NMA can inhibit iNOS-dependent NO production by preventing iNOS dimerization and destabilizing its dimeric form.
Collapse
Affiliation(s)
- Kyu-Sun Lee
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Gahman TC, Herbert MR, Lang H, Thayer A, Symons KT, Nguyen PM, Massari ME, Dozier S, Zhang Y, Sablad M, Rao TS, Noble SA, Shiau AK, Hassig CA. Identification and SAR of selective inducible nitric oxide synthase (iNOS) dimerization inhibitors. Bioorg Med Chem Lett 2011; 21:6888-94. [DOI: 10.1016/j.bmcl.2011.08.112] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Revised: 08/20/2011] [Accepted: 08/25/2011] [Indexed: 11/25/2022]
|