1
|
Salvadori L, Paiella M, Castiglioni B, Belladonna ML, Manenti T, Ercolani C, Cornioli L, Clemente N, Scircoli A, Sardella R, Tensi L, Astolfi A, Barreca ML, Chiappalupi S, Gentili G, Bosetti M, Sorci G, Filigheddu N, Riuzzi F. Equisetum arvense standardized dried extract hinders age-related osteosarcopenia. Biomed Pharmacother 2024; 174:116517. [PMID: 38574619 DOI: 10.1016/j.biopha.2024.116517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/20/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024] Open
Abstract
Age-associated osteosarcopenia is an unresolved syndrome characterized by the concomitant loss of bone (osteopenia) and skeletal muscle (sarcopenia) tissues increasing falls, immobility, morbidity, and mortality. Unbalanced resorption of bone in the remodeling process and excessive protein breakdown, especially fast type II myosin heavy chain (MyHC-II) isoform and myofiber metabolic shift, are the leading causes of bone and muscle deterioration in the elderly, respectively. Equisetum arvense (EQ) is a plant traditionally recommended for many pathological conditions due to its anti-inflammatory properties. Thus, considering that a chronic low-grade inflammatory state predisposes to both osteoporosis and sarcopenia, we tested a standardized hydroalcoholic extract of EQ in in vitro models of muscle atrophy [C2C12 myotubes treated with proinflammatory cytokines (TNFα/IFNγ), excess glucocorticoids (dexamethasone), or the osteokine, receptor activator of nuclear factor kappa-B ligand (RANKL)] and osteoclastogenesis (RAW 264.7 cells treated with RANKL). We found that EQ counteracted myotube atrophy, blunting the activity of several pathways depending on the applied stimulus, and reduced osteoclast formation and activity. By in silico target fishing, IKKB-dependent nuclear factor kappa-B (NF-κB) inhibition emerges as a potential common mechanism underlying EQ's anti-atrophic effects. Consumption of EQ (500 mg/kg/day) by pre-geriatric C57BL/6 mice for 3 months translated into: i) maintenance of muscle mass and performance; ii) restrained myofiber oxidative shift; iii) slowed down age-related modifications in osteoporotic bone, significantly preserving trabecular connectivity density; iv) reduced muscle- and spleen-related inflammation. EQ can preserve muscle functionality and bone remodeling during aging, potentially valuable as a natural treatment for osteosarcopenia.
Collapse
Affiliation(s)
- Laura Salvadori
- Department of Translational Medicine, University of Piemonte Orientale, Novara 28100, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy
| | - Martina Paiella
- Department of Translational Medicine, University of Piemonte Orientale, Novara 28100, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy
| | - Beatrice Castiglioni
- Department Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | | | | | | | - Luca Cornioli
- Laboratori Biokyma srl, Anghiari, Arezzo 52031, Italy
| | - Nausicaa Clemente
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara 28100, Italy
| | - Andrea Scircoli
- Department of Translational Medicine, University of Piemonte Orientale, Novara 28100, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy
| | - Roccaldo Sardella
- Department of Pharmaceutical Sciences, University of Perugia, Perugia 06123, Italy
| | - Leonardo Tensi
- Department of Pharmaceutical Sciences, University of Perugia, Perugia 06123, Italy
| | - Andrea Astolfi
- Department of Pharmaceutical Sciences, University of Perugia, Perugia 06123, Italy
| | | | - Sara Chiappalupi
- Interuniversity Institute of Myology (IIM), Perugia 06132, Italy; Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Giulia Gentili
- Interuniversity Institute of Myology (IIM), Perugia 06132, Italy; Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Michela Bosetti
- Department Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Guglielmo Sorci
- Interuniversity Institute of Myology (IIM), Perugia 06132, Italy; Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Nicoletta Filigheddu
- Department of Translational Medicine, University of Piemonte Orientale, Novara 28100, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy
| | - Francesca Riuzzi
- Interuniversity Institute of Myology (IIM), Perugia 06132, Italy; Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy.
| |
Collapse
|
2
|
Guan F, Wang Q, Bao Y, Chao Y. Anti-rheumatic effect of quercetin and recent developments in nano formulation. RSC Adv 2021; 11:7280-7293. [PMID: 35423269 PMCID: PMC8695102 DOI: 10.1039/d0ra08817j] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Quercetin is a potential anti-rheumatoid drug. Nano formulation strategies could improve its solubility and efficacy.
Collapse
Affiliation(s)
- Feng Guan
- School of Pharmacy
- Heilongjiang University of Chinese Medicine
- Harbin 150040
- P. R. China
| | - Qi Wang
- Norwich Medical School
- University of East Anglia
- Norwich NR4 7UQ
- UK
| | - Yongping Bao
- Norwich Medical School
- University of East Anglia
- Norwich NR4 7UQ
- UK
| | - Yimin Chao
- School of Chemistry
- University of East Anglia
- Norwich NR4 7TJ
- UK
| |
Collapse
|
3
|
Amanzadeh E, Esmaeili A, Abadi REN, Kazemipour N, Pahlevanneshan Z, Beheshti S. Quercetin conjugated with superparamagnetic iron oxide nanoparticles improves learning and memory better than free quercetin via interacting with proteins involved in LTP. Sci Rep 2019; 9:6876. [PMID: 31053743 PMCID: PMC6499818 DOI: 10.1038/s41598-019-43345-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 04/23/2019] [Indexed: 01/20/2023] Open
Abstract
Biomedical application of quercetin (QT) as an effective flavonoid has limitations due to its low bioavailability. Superparamagnetic iron oxide nanoparticle (SPION) is a novel drug delivery system that enhances the bioavailability of quercetin. The effect of short time usage of quercetin on learning and memory function and its signaling pathways in the healthy rat is not well understood. The aim of this study was to investigate the effect of free quercetin and in conjugation with SPION on learning and memory in healthy rats and to find quercetin target proteins involved in learning and memory using Morris water maze (MWM) and computational methods respectively. Results of MWM show an improvement in learning and memory of rats treated with either quercetin or QT-SPION. Better learning and memory functions using QT-SPION reveal increased bioavailability of quercetin. Comparative molecular docking studies show the better binding affinity of quercetin to RSK2, MSK1, CytC, Cdc42, Apaf1, FADD, CRK proteins. Quercetin in comparison to specific inhibitors of each protein also demonstrates a better QT binding affinity. This suggests that quercetin binds to proteins leading to prevent neural cell apoptosis and improves learning and memory. Therefore, SPIONs could increase the bioavailability of quercetin and by this way improve learning and memory.
Collapse
Affiliation(s)
- Elnaz Amanzadeh
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Abolghasem Esmaeili
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran.
| | | | - Nasrin Kazemipour
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Zari Pahlevanneshan
- Department of Chemistry, Catalysis Division, University of Isfahan, Isfahan, Iran
| | - Siamak Beheshti
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| |
Collapse
|
4
|
Paul A, Edwards J, Pepper C, Mackay S. Inhibitory-κB Kinase (IKK) α and Nuclear Factor-κB (NFκB)-Inducing Kinase (NIK) as Anti-Cancer Drug Targets. Cells 2018; 7:E176. [PMID: 30347849 PMCID: PMC6210445 DOI: 10.3390/cells7100176] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/15/2018] [Accepted: 10/17/2018] [Indexed: 12/23/2022] Open
Abstract
The cellular kinases inhibitory-κB kinase (IKK) α and Nuclear Factor-κB (NF-κB)-inducing kinase (NIK) are well recognised as key central regulators and drivers of the non-canonical NF-κB cascade and as such dictate the initiation and development of defined transcriptional responses associated with the liberation of p52-RelB and p52-p52 NF-κB dimer complexes. Whilst these kinases and downstream NF-κB complexes transduce pro-inflammatory and growth stimulating signals that contribute to major cellular processes, they also play a key role in the pathogenesis of a number of inflammatory-based conditions and diverse cancer types, which for the latter may be a result of background mutational status. IKKα and NIK, therefore, represent attractive targets for pharmacological intervention. Here, specifically in the cancer setting, we reflect on the potential pathophysiological role(s) of each of these kinases, their associated downstream signalling outcomes and the stimulatory and mutational mechanisms leading to their increased activation. We also consider the downstream coordination of transcriptional events and phenotypic outcomes illustrative of key cancer 'Hallmarks' that are now increasingly perceived to be due to the coordinated recruitment of both NF-κB-dependent as well as NF-κB⁻independent signalling. Furthermore, as these kinases regulate the transition from hormone-dependent to hormone-independent growth in defined tumour subsets, potential tumour reactivation and major cytokine and chemokine species that may have significant bearing upon tumour-stromal communication and tumour microenvironment it reiterates their potential to be drug targets. Therefore, with the emergence of small molecule kinase inhibitors targeting each of these kinases, we consider medicinal chemistry efforts to date and those evolving that may contribute to the development of viable pharmacological intervention strategies to target a variety of tumour types.
Collapse
Affiliation(s)
- Andrew Paul
- Strathclyde Institute of Pharmacy and Biomedical Sciences, 161 Cathedral Street, University of Strathclyde, Glasgow G4 0NR, UK.
| | - Joanne Edwards
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1QH, UK.
| | - Christopher Pepper
- Brighton and Sussex Medical School, University of Sussex, Brighton BN1 9PX, UK.
| | - Simon Mackay
- Strathclyde Institute of Pharmacy and Biomedical Sciences, 161 Cathedral Street, University of Strathclyde, Glasgow G4 0NR, UK.
| |
Collapse
|
5
|
Ho TY, Li CC, Lo HY, Chen FY, Hsiang CY. Corn Silk Extract and Its Bioactive Peptide Ameliorated Lipopolysaccharide-Induced Inflammation in Mice via the Nuclear Factor-κB Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:759-768. [PMID: 28064493 DOI: 10.1021/acs.jafc.6b03327] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Bioactive peptides derived from foods have shown beneficial anti-inflammatory potential. Inhibitory κB kinase-β (IKKβ) plays a crucial role in the activation of nuclear factor-κB (NF-κB), a transcription factor involved in inflammation. Here we applied proteomic and bioinformatics approaches to identify anti-inflammatory peptides that target IKKβ from corn silk. Corn silk extract significantly suppressed lipopolysaccharide (LPS)-induced NF-κB activities [(1.7 ± 0.2)-fold vs (3.0 ± 0.6)-fold, p < 0.05] in cells. Trypsin hydrolysate of corn silk also suppressed LPS-induced NF-κB activities [(1.1 ± 0.3)-fold vs 3.3 ± 0.5 fold, p < 0.01]. In addition, both corn silk extract and trypsin hydrolysate significantly inhibited LPS-induced interleukin-1β (IL-1β) production by 58.3 ± 4.5 and 55.1 ± 7.4%, respectively. A novel peptide, FK2, docked into the ATP-binding pocket of IKKβ, was further identified from trypsin hydrolysis of corn silk. FK2 inhibited IKKβ activities, IκB phosphorylation, and subsequent NF-κB activation [(2.3 ± 0.4)-fold vs (5.5 ± 0.4)-fold, p < 0.001]. Moreover, FK2 significantly reduced NF-κB-driven luminescent signals in organs by 5-11-fold and suppressed LPS-induced NF-κB activities and IL-β production in tissues. In conclusion, our findings indicated that corn silk displayed anti-inflammatory abilities. In addition, we first identified an anti-inflammatory peptide FK2 from corn silk. Moreover, the anti-inflammatory effect of FK2 might be through IKKβ-NF-κB signaling pathways.
Collapse
Affiliation(s)
- Tin-Yun Ho
- Graduate Institute of Chinese Medicine, China Medical University , Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University , Taichung, Taiwan
| | - Chia-Cheng Li
- Graduate Institute of Chinese Medicine, China Medical University , Taichung, Taiwan
| | - Hsin-Yi Lo
- Graduate Institute of Chinese Medicine, China Medical University , Taichung, Taiwan
| | - Feng-Yuan Chen
- Graduate Institute of Chinese Medicine, China Medical University , Taichung, Taiwan
| | - Chien-Yun Hsiang
- Department of Microbiology, China Medical University , Taichung, Taiwan
| |
Collapse
|
6
|
Heim KC, Angers P, Léonhart S, Ritz BW. Anti-Inflammatory and Neuroactive Properties of Selected Fruit Extracts. J Med Food 2012; 15:851-4. [DOI: 10.1089/jmf.2011.0265] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Kelly C. Heim
- Pure Encapsulations, Inc., Sudbury, Massachusetts, USA
| | - Paul Angers
- Center of Horticulture Research, Laval University, Quebec City, Quebec, Canada
| | | | - Barry W. Ritz
- Atrium Innovations, Inc., Chadds Ford, Pennsylvania, USA
| |
Collapse
|
7
|
Kalia M, Kukol A. Structure and dynamics of the kinase IKK-β--A key regulator of the NF-kappa B transcription factor. J Struct Biol 2011; 176:133-42. [PMID: 21820058 DOI: 10.1016/j.jsb.2011.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 07/19/2011] [Accepted: 07/20/2011] [Indexed: 12/29/2022]
Abstract
The inhibitor κB kinase-β (IKK-β) phosphorylates the NF-κB inhibitor protein IκB leading to the translocation of the transcription factor NF-κB to the nucleus. The transcription factor NF-κB and consequently IKK-β are central to signal transduction pathways of mammalian cells. The purpose of this research was to develop a 3D structural model of the IKK-β kinase domain with its ATP cofactor and investigate its dynamics and ligand binding potential. Through a combination of comparative modelling and simulated heating/annealing molecular dynamics (SAMD) simulation in explicit water the model accuracy could be substantially improved compared to comparative modelling on its own as shown by model validation measures. The structure revealed the details of ATP/Mg(2+) binding indicating hydrophobic interactions with the adenine base and a significant contribution of Mg(2+) as a bridge between ATP phosphate groups and negatively charged side chains. The molecular dynamics trajectories of the ATP-bound and free enzyme showed two conformations in each case, which contributed to the majority of the trajectory. The ATP-free enzyme revealed a novel binding site distant from the ATP binding site that was not encountered in the ATP bound enzyme. Based on the overall structural flexibility, it is suggested that a truncated version of the kinase domain from Ala14 to Leu265 should be subjected to crystallisation trials. The 3D structure of this enzyme will enable rational design of new ligands and analysis of protein-protein interactions. Furthermore, our results may provide a new impetus for wet-lab based structural investigation focussing on a truncated kinase domain.
Collapse
Affiliation(s)
- Munishikha Kalia
- School of Life Sciences, University of Hertfordshire, Hatfield AL10 9AB, United Kingdom
| | | |
Collapse
|
8
|
Sala E, Guasch L, Iwaszkiewicz J, Mulero M, Salvadó MJ, Pinent M, Zoete V, Grosdidier A, Garcia-Vallvé S, Michielin O, Pujadas G. Identification of human IKK-2 inhibitors of natural origin (part I): modeling of the IKK-2 kinase domain, virtual screening and activity assays. PLoS One 2011; 6:e16903. [PMID: 21390216 PMCID: PMC3044726 DOI: 10.1371/journal.pone.0016903] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 01/14/2011] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Their large scaffold diversity and properties, such as structural complexity and drug similarity, form the basis of claims that natural products are ideal starting points for drug design and development. Consequently, there has been great interest in determining whether such molecules show biological activity toward protein targets of pharmacological relevance. One target of particular interest is hIKK-2, a serine-threonine protein kinase belonging to the IKK complex that is the primary component responsible for activating NF-κB in response to various inflammatory stimuli. Indeed, this has led to the development of synthetic ATP-competitive inhibitors for hIKK-2. Therefore, the main goals of this study were (a) to use virtual screening to identify potential hIKK-2 inhibitors of natural origin that compete with ATP and (b) to evaluate the reliability of our virtual-screening protocol by experimentally testing the in vitro activity of selected natural-product hits. METHODOLOGY/PRINCIPAL FINDINGS We thus predicted that 1,061 out of the 89,425 natural products present in the studied database would inhibit hIKK-2 with good ADMET properties. Notably, when these 1,061 molecules were merged with the 98 synthetic hIKK-2 inhibitors used in this study and the resulting set was classified into ten clusters according to chemical similarity, there were three clusters that contained only natural products. Five molecules from these three clusters (for which no anti-inflammatory activity has been previously described) were then selected for in vitro activity testing, in which three out of the five molecules were shown to inhibit hIKK-2. CONCLUSIONS/SIGNIFICANCE We demonstrated that our virtual-screening protocol was successful in identifying lead compounds for developing new inhibitors for hIKK-2, a target of great interest in medicinal chemistry. Additionally, all the tools developed during the current study (i.e., the homology model for the hIKK-2 kinase domain and the pharmacophore) will be made available to interested readers upon request.
Collapse
Affiliation(s)
- Esther Sala
- Grup de Recerca en Nutrigenòmica, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, Campus de Sescelades, Tarragona, Catalonia, Spain
| | - Laura Guasch
- Grup de Recerca en Nutrigenòmica, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, Campus de Sescelades, Tarragona, Catalonia, Spain
| | - Justyna Iwaszkiewicz
- Molecular Modeling Group, Swiss Institute of Bioinformatics, Quartier UNIL-Sorge, Lausanne, Switzerland
| | - Miquel Mulero
- Grup de Recerca en Nutrigenòmica, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, Campus de Sescelades, Tarragona, Catalonia, Spain
| | - Maria-Josepa Salvadó
- Grup de Recerca en Nutrigenòmica, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, Campus de Sescelades, Tarragona, Catalonia, Spain
| | - Montserrat Pinent
- Grup de Recerca en Nutrigenòmica, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, Campus de Sescelades, Tarragona, Catalonia, Spain
| | - Vincent Zoete
- Molecular Modeling Group, Swiss Institute of Bioinformatics, Quartier UNIL-Sorge, Lausanne, Switzerland
| | - Aurélien Grosdidier
- Molecular Modeling Group, Swiss Institute of Bioinformatics, Quartier UNIL-Sorge, Lausanne, Switzerland
| | - Santiago Garcia-Vallvé
- Grup de Recerca en Nutrigenòmica, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, Campus de Sescelades, Tarragona, Catalonia, Spain
- Centre Tecnològic de Nutrició i Salut, Reus, Catalonia, Spain
| | - Olivier Michielin
- Molecular Modeling Group, Swiss Institute of Bioinformatics, Quartier UNIL-Sorge, Lausanne, Switzerland
| | - Gerard Pujadas
- Grup de Recerca en Nutrigenòmica, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, Campus de Sescelades, Tarragona, Catalonia, Spain
- Centre Tecnològic de Nutrició i Salut, Reus, Catalonia, Spain
| |
Collapse
|
9
|
Avila CM, Lopes AB, Gonçalves AS, da Silva LL, Romeiro NC, Miranda ALP, Sant'Anna CMR, Barreiro EJ, Fraga CAM. Structure-based design and biological profile of (E)-N-(4-Nitrobenzylidene)-2-naphthohydrazide, a novel small molecule inhibitor of IκB kinase-β. Eur J Med Chem 2011; 46:1245-53. [PMID: 21334796 DOI: 10.1016/j.ejmech.2011.01.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 01/14/2011] [Accepted: 01/25/2011] [Indexed: 01/04/2023]
Abstract
In this study, we describe the rational design, molecular modeling and pharmacological profile of a novel IKK-β inhibitor (E)-N-(4-nitrobenzylidene)-2-naphthohydrazide (LASSBio-1524). The design based on the IKK-β active site, and a privileged structure template yielded a novel IKK-β inhibitor scaffold with significant selectivity over IKK-α and CHK2, as assessed by an in vitro kinase assay. For a better understanding of the structural requirements of IKK-β inhibition, molecular dynamics simulations of LASSBio-1524 (3) were performed. The NAH derivative LASSBio-1524 (3), was able to suppress arachidonic acid-induced edema formation in a dose-dependent manner, demonstrating an in vivo anti-inflammatory effect. The molecular architecture of this novel, low-molecular weight IKK-β inhibitor is encouraging for further lead optimization toward the development of innovative anti-inflammatory drug candidates.
Collapse
Affiliation(s)
- Carolina M Avila
- Laboratório de Avaliação e Síntese de Substâncias Bioativas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, PO Box 68023, 21941-902 Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Lauria A, Ippolito M, Fazzari M, Tutone M, Di Blasi F, Mingoia F, Almerico AM. IKK-beta inhibitors: an analysis of drug-receptor interaction by using molecular docking and pharmacophore 3D-QSAR approaches. J Mol Graph Model 2010; 29:72-81. [PMID: 20537930 DOI: 10.1016/j.jmgm.2010.04.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2009] [Revised: 04/28/2010] [Accepted: 04/30/2010] [Indexed: 11/29/2022]
Abstract
The IKK kinases family represents a thrilling area of research because of its importance in regulating the activity of NF-kB transcription factors. The discovery of the central role played by IKK-beta in the activation of transcription in response to apoptotic or inflammatory stimuli allowed to considerate its modulation as a promising tool for the treatment of chronic inflammation and cancer. To date, several IKK-beta inhibitors have been discovered and tested. In this work, an analysis of the interactions between different classes of inhibitors and their biological target was performed, through the application of Molecular Docking and Pharmacophore/3D-QSAR approaches to a set of 141 inhibitors included in the Binding Database. In order to overcome the difficulty due to the lack of crystallographic data for IKK-beta, a homology model of this protein has been built and validated. The results allowed to study in depth the structural bases for the interaction of each family of inhibitors and provided clues for further modifications, with the aim of improving the activity and selectivity of designed drugs targeting this enzyme.
Collapse
Affiliation(s)
- Antonino Lauria
- Dipartimento Farmacochimico, Tossicologico e Biologico, Università di Palermo, Via Archirafi 32, Palermo, Italy.
| | | | | | | | | | | | | |
Collapse
|