1
|
Bhargava S, Kulkarni R, Dewangan B, Kulkarni N, Jiaswar C, Kumar K, Kumar A, Bodhe PR, Kumar H, Sahu B. Microtubule stabilising peptides: new paradigm towards management of neuronal disorders. RSC Med Chem 2023; 14:2192-2205. [PMID: 37974959 PMCID: PMC10650357 DOI: 10.1039/d3md00012e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/09/2023] [Indexed: 11/19/2023] Open
Abstract
Neuronal cells made of soma, axon, and dendrites are highly compartmentalized and possess a specialized transport system that can convey long-distance electrical signals for the cross-talk. The transport system is made up of microtubule (MT) polymers and MT-binding proteins. MTs play vital and diverse roles in various cellular processes. Therefore, defects and dysregulation of MTs and their binding proteins lead to many neurological disorders as exemplified by Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, Huntington's disease, and many others. MT-stabilising agents (MSAs) altering the MT-associated protein connections have shown great potential for several neurodegenerative disorders. Peptides are an important class of molecules with high specificity, biocompatibility and are devoid of side effects. In the past, peptides have been explored in various neuronal disorders as therapeutics. Davunetide, a MT-stabilising octapeptide, has entered into phase II clinical trials for schizophrenia. Numerous examples of peptides emerging as MSAs reflect the emergence of a new paradigm for peptides which can be explored further as drug candidates for neuronal disorders. Although small molecule-based MSAs have been reviewed in the past, there is no systematic review in recent years focusing on peptides as MSAs apart from davunetide in 2013. Therefore, a systematic updated review on MT stabilising peptides may shed light on many hidden aspects and enable researchers to develop new therapies for diseases related to the CNS. In this review we have summarised the recent examples of peptides as MSAs.
Collapse
Affiliation(s)
- Shubhangi Bhargava
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Riya Kulkarni
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Bhaskar Dewangan
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Neeraj Kulkarni
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Chirag Jiaswar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Kunal Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Amit Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Praveen Reddy Bodhe
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Ahmedabad India
| | - Bichismita Sahu
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad India
| |
Collapse
|
2
|
Abstract
Targeted protein degradation (TPD) has emerged as the most promising approach for the specific knockdown of disease-associated proteins and is achieved by exploiting the cellular quality control machinery. TPD technologies are highly advantageous in overcoming drug resistance as they degrade the whole target protein. Microtubules play important roles in many cellular processes and are among the oldest and most well-established targets for tumor chemotherapy. However, the development of drug resistance, risk of hypersensitivity reactions, and intolerable toxicities severely restrict the clinical applications of microtubule-targeting agents (MTAs). Microtubule degradation agents (MDgAs) operate via completely different mechanisms compared with traditional MTAs and are capable of overcoming drug resistance. The emergence of MDgAs has expanded the scope of TPD and provided new avenues for the discovery of tubulin-targeted drugs. Herein, we summarized the development of MDgAs, and discussed their degradation mechanisms, mechanisms of action on the binding sites, potential opportunities, and challenges.
Collapse
Affiliation(s)
- Chufeng Zhang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Min Zhao
- Laboratory of Metabolomics and Drug-Induced Liver Injury, Department of Gastroenterology & Hepatology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Guan Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yong Li
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
3
|
El-Atawy MA, Alshaye NA, Elrubi N, Hamed EA, Omar AZ. Pyrimidines-Based Heterocyclic Compounds: Synthesis, Cytoxicity Evaluation and Molecular Docking. Molecules 2022; 27:molecules27154912. [PMID: 35956864 PMCID: PMC9370056 DOI: 10.3390/molecules27154912] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 11/23/2022] Open
Abstract
A variety of structurally different pyrimidines were synthesized. Elemental analysis, FT-IR, 1H NMR, and 13C NMR spectroscopy were used to confirm the chemical structures of all prepared compounds. The synthesized pyrimidines were screened against the growth of five human cancer cell lines (prostate carcinoma PC3, liver carcinoma HepG-2, human colon cancer HCT-116, human breast cancer MCF-7, human lung cancer A-549), and normal human lung fibroblasts (MRC-5) using MTT assay. Most of the screened pyrimidines have anti-proliferative activity on the growth of the PC3 cell line. Compounds 3b and 3d were more potent than the reference vinblastine sulfate (~2 to 3 × fold) and they can be considered promising leads for treating prostate cancer disease. Moreover, the screened compounds 3b, 3f, 3g, 3h, and 5 were assessed according to the values of their selectivity index (SI) and were found to be more selective and safer than vinblastine sulfate. Furthermore, using in silico computational tools, the physicochemical properties of all pyrimidine ligands were assessed, and the synthesized compounds fall within the criteria of RO5, thus having the potential to be orally bioavailable.
Collapse
Affiliation(s)
- Mohamed A. El-Atawy
- Chemistry Department, Faculty of Science, Taibah University, Yanbu 46423, Saudi Arabia
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (N.E.); (E.A.H.)
- Correspondence: (M.A.E.-A.); (A.Z.O.); Tel.: +966-569-191-532 (M.A.E.-A.); +20-111-136-1784 (A.Z.O.)
| | - Najla A. Alshaye
- Department of Chemistry, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | - Nada Elrubi
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (N.E.); (E.A.H.)
| | - Ezzat A. Hamed
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (N.E.); (E.A.H.)
| | - Alaa Z. Omar
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (N.E.); (E.A.H.)
- Correspondence: (M.A.E.-A.); (A.Z.O.); Tel.: +966-569-191-532 (M.A.E.-A.); +20-111-136-1784 (A.Z.O.)
| |
Collapse
|
4
|
Bose D, Roy L, Chatterjee S. Peptide therapeutics in the management of metastatic cancers. RSC Adv 2022; 12:21353-21373. [PMID: 35975072 PMCID: PMC9345020 DOI: 10.1039/d2ra02062a] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/26/2022] [Indexed: 11/21/2022] Open
Abstract
Cancer remains a leading health concern threatening lives of millions of patients worldwide. Peptide-based drugs provide a valuable alternative to chemotherapeutics as they are highly specific, cheap, less toxic and easier to synthesize compared to other drugs. In this review, we have discussed various modes in which peptides are being used to curb cancer. Our review highlights specially the various anti-metastatic peptide-based agents developed by targeting a plethora of cellular factors. Herein we have given a special focus on integrins as targets for peptide drugs, as these molecules play key roles in metastatic progression. The review also discusses use of peptides as anti-cancer vaccines and their efficiency as drug-delivery tools. We hope this work will give the reader a clear idea of the mechanisms of peptide-based anti-cancer therapeutics and encourage the development of superior drugs in the future.
Collapse
Affiliation(s)
- Debopriya Bose
- Department of Biophysics Bose Institute Unified Academic Campus EN 80, Sector V, Bidhan Nagar Kolkata 700091 WB India
| | - Laboni Roy
- Department of Biophysics Bose Institute Unified Academic Campus EN 80, Sector V, Bidhan Nagar Kolkata 700091 WB India
| | - Subhrangsu Chatterjee
- Department of Biophysics Bose Institute Unified Academic Campus EN 80, Sector V, Bidhan Nagar Kolkata 700091 WB India
| |
Collapse
|
5
|
Relitti N, Saraswati AP, Carullo G, Papa A, Monti A, Benedetti R, Passaro E, Brogi S, Calderone V, Butini S, Gemma S, Altucci L, Campiani G, Doti N. Design and Synthesis of New Oligopeptidic Parvulin Inhibitors. ChemMedChem 2022; 17:e202200050. [PMID: 35357776 PMCID: PMC9321596 DOI: 10.1002/cmdc.202200050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/28/2022] [Indexed: 11/12/2022]
Abstract
Pin1 catalyzes the cis-trans isomerization of pThr-Pro or pSer-Pro amide bonds of different proteins involved in several physio/pathological processes. In this framework, recent research activity is directed towards the identification of new selective Pin1 inhibitors. Here, we developed a set ( 5a - p ) of peptide-based Pin1 inhibitors. Direct-binding experiments allowed the identification of the peptide-based inhibitor 5k as a potent ligand of Pin1. Notably, 5k binds Pin1 with a higher affinity compared to Pin4. The comparative analysis of molecular models of Pin1 and Pin4 with the selected compound, gave a rational explanation of the biochemical activity, and pinpointed the chemical elements that, if opportunely modified, may further improve inhibitory potency, pharmacological properties and selectivity of future peptide-based Parvulin inhibitors. Since 5k showed a limited cell penetration and no antiproliferative activity, it was conjugated to a polyarginine stretch, known to promote cell penetration of peptides, to obtain R8-5k derivative, which displayed an anti-proliferative effect on cancer cell lines compared to non-tumor cells. The effect of R8 on cell proliferation was also investigated. This work doubts the application of the R8 strategy for the development of cell penetrating antiproliferative peptides since it is not inert.
Collapse
Affiliation(s)
- Nicola Relitti
- University of Siena: Universita degli Studi di Siena, DBCF, ITALY
| | | | - Gabriele Carullo
- University of Siena: Universita degli Studi di Siena, DBCF, 2, Aldo Moro, 53100 Siena Italy, 53100, Siena, ITALY
| | - Alessandro Papa
- University of Siena: Universita degli Studi di Siena, DBCF, ITALY
| | | | - Rosaria Benedetti
- University of Campania Luigi Vanvitelli: Universita degli Studi della Campania Luigi Vanvitelli, Medicine, ITALY
| | - Eugenia Passaro
- University of Pisa Department of Pharmaceutical Sciences: Universita degli Studi di Pisa Dipartimento di Farmacia, Pharmacy, ITALY
| | - Simone Brogi
- University of Pisa Department of Pharmaceutical Sciences: Universita degli Studi di Pisa Dipartimento di Farmacia, Pharmacy, ITALY
| | - Vincenzo Calderone
- University of Pisa Department of Pharmaceutical Sciences: Universita degli Studi di Pisa Dipartimento di Farmacia, Pharmacy, ITALY
| | - Stefania Butini
- University of Siena: Universita degli Studi di Siena, DBCF, ITALY
| | - Sandra Gemma
- University of Siena: Universita degli Studi di Siena, DBCF, ITALY
| | - Lucia Altucci
- University of Campania Luigi Vanvitelli: Universita degli Studi della Campania Luigi Vanvitelli, Medicine, ITALY
| | - Giuseppe Campiani
- Universita degli Studi di Siena, Dipartimento di Biotecnologie, Via Aldo Moro 2, 53100, Siena, ITALY
| | - Nunzianna Doti
- CNR: Consiglio Nazionale delle Ricerche, Bioimaging, ITALY
| |
Collapse
|
6
|
The application progress of peptides in drug delivery systems in the past decade. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
7
|
Gabr MT, Brogi S. MicroRNA-Based Multitarget Approach for Alzheimer's Disease: Discovery of the First-In-Class Dual Inhibitor of Acetylcholinesterase and MicroRNA-15b Biogenesis. J Med Chem 2020; 63:9695-9704. [PMID: 32787143 DOI: 10.1021/acs.jmedchem.0c00756] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The multitarget approach in drug design is a powerful strategy in tackling the multifactorial nature of Alzheimer's disease (AD). Herein, we report a novel strategy in the design of multitargeted therapeutics for AD through dual inhibition of acetylcholinesterase (AChE) and microRNA-15b biogenesis. We performed high-throughput screening (HTS) of a chemical library to identify binders of mircoRNA-15b which is identified as a biomarker and potential therapeutic target of AD. The hits from HTS were further screened for their AChE inhibitory activity, the most widely investigated target for the development of AD therapeutics. MG-6267 was identified as the first dual inhibitor of AChE and microRNA-15b biogenesis. Cellular assays revealed the superiority of MG-6267 to single-targeted inhibitors of AChE and microRNA-15b in protecting SH-SY5Y neuroblastoma cells from amyloid-beta (Aβ)-induced cytotoxicity. This work paves the way for future research efforts aiming at the development of microRNA-based multitargeted therapeutics for AD.
Collapse
Affiliation(s)
- Moustafa T Gabr
- Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy
| |
Collapse
|
8
|
Multitarget Therapeutic Strategies for Alzheimer's Disease: Review on Emerging Target Combinations. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5120230. [PMID: 32714977 PMCID: PMC7354643 DOI: 10.1155/2020/5120230] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/12/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases represent nowadays one of the major health problems. Despite the efforts made to unveil the mechanism leading to neurodegeneration, it is still not entirely clear what triggers this phenomenon and what allows its progression. Nevertheless, it is accepted that neurodegeneration is a consequence of several detrimental processes, such as protein aggregation, oxidative stress, and neuroinflammation, finally resulting in the loss of neuronal functions. Starting from these evidences, there has been a wide search for novel agents able to address more than a single event at the same time, the so-called multitarget-directed ligands (MTDLs). These compounds originated from the combination of different pharmacophoric elements which endowed them with the ability to interfere with different enzymatic and/or receptor systems, or to exert neuroprotective effects by modulating proteins and metal homeostasis. MTDLs have been the focus of the latest strategies to discover a new treatment for Alzheimer's disease (AD), which is considered the most common form of dementia characterized by neurodegeneration and cognitive dysfunctions. This review is aimed at collecting the latest and most interesting target combinations for the treatment of AD, with a detailed discussion on new agents with favorable in vitro properties and on optimized structures that have already been assessed in vivo in animal models of dementia.
Collapse
|
9
|
Inaba H, Nagata M, Miyake KJ, Kabir AMR, Kakugo A, Sada K, Matsuura K. Cyclic Tau-derived peptides for stabilization of microtubules. Polym J 2020. [DOI: 10.1038/s41428-020-0356-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
10
|
Brogi S, Sirous H, Calderone V, Chemi G. Amyloid β fibril disruption by oleuropein aglycone: long-time molecular dynamics simulation to gain insight into the mechanism of action of this polyphenol from extra virgin olive oil. Food Funct 2020; 11:8122-8132. [DOI: 10.1039/d0fo01511c] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Insight into the mechanism of action of oleuropein aglycone as a potent anti-amyloidogenic agent.
Collapse
Affiliation(s)
- Simone Brogi
- Department of Pharmacy
- University of Pisa
- 56126 Pisa
- Italy
| | - Hajar Sirous
- Bioinformatics Research Center
- School of Pharmacy and Pharmaceutical Sciences
- Isfahan University of Medical Sciences
- 81746-73461 Isfahan
- Iran
| | | | - Giulia Chemi
- Wellcome Centre for Anti-Infectives Research
- Drug Discovery Unit
- Division of Biological Chemistry and Drug Discovery
- University of Dundee
- DD1 5EH Dundee
| |
Collapse
|
11
|
Synthesis, biological evaluation and molecular modeling of novel selective COX-2 inhibitors: sulfide, sulfoxide, and sulfone derivatives of 1,5-diarylpyrrol-3-substituted scaffold. Bioorg Med Chem 2019; 27:115045. [DOI: 10.1016/j.bmc.2019.115045] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 12/27/2022]
|
12
|
Sirous H, Chemi G, Gemma S, Butini S, Debyser Z, Christ F, Saghaie L, Brogi S, Fassihi A, Campiani G, Brindisi M. Identification of Novel 3-Hydroxy-pyran-4-One Derivatives as Potent HIV-1 Integrase Inhibitors Using in silico Structure-Based Combinatorial Library Design Approach. Front Chem 2019; 7:574. [PMID: 31457006 PMCID: PMC6700280 DOI: 10.3389/fchem.2019.00574] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 07/29/2019] [Indexed: 11/16/2022] Open
Abstract
We describe herein the development and experimental validation of a computational protocol for optimizing a series of 3-hydroxy-pyran-4-one derivatives as HIV integrase inhibitors (HIV INIs). Starting from a previously developed micromolar inhibitors of HIV integrase (HIV IN), we performed an in-depth investigation based on an in silico structure-based combinatorial library designing approach. This method allowed us to combine a combinatorial library design and side chain hopping with Quantum Polarized Ligand Docking (QPLD) studies and Molecular Dynamics (MD) simulation. The combinatorial library design allowed the identification of the best decorations for our promising scaffold. The resulting compounds were assessed by the mentioned QPLD methodology using a homology model of full-length binary HIV IN/DNA for retrieving the best performing compounds acting as HIV INIs. Along with the prediction of physico-chemical properties, we were able to select a limited number of drug-like compounds potentially displaying potent HIV IN inhibition. From this final set, based on the synthetic accessibility, we further shortlisted three representative compounds for the synthesis. The compounds were experimentally assessed in vitro for evaluating overall HIV-1 IN inhibition, HIV-1 IN strand transfer activity inhibition, HIV-1 activity inhibition and cellular toxicity. Gratifyingly, all of them showed relevant inhibitory activity in the in vitro tests along with no toxicity. Among them HPCAR-28 represents the most promising compound as potential anti-HIV agent, showing inhibitory activity against HIV IN in the low nanomolar range, comparable to that found for Raltegravir, and relevant potency in inhibiting HIV-1 replication and HIV-1 IN strand transfer activity. In summary, our results outline HPCAR-28 as a useful optimized hit for the potential treatment of HIV-1 infection by targeting HIV IN.
Collapse
Affiliation(s)
- Hajar Sirous
- Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Giulia Chemi
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, Siena, Italy
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, Siena, Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, Siena, Italy
| | - Zeger Debyser
- Molecular Medicine, K.U. Leuven and IRC KULAK, Leuven, Belgium
| | - Frauke Christ
- Molecular Medicine, K.U. Leuven and IRC KULAK, Leuven, Belgium
| | - Lotfollah Saghaie
- Department of Medicinal Chemistry, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Afshin Fassihi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, Siena, Italy
| | - Margherita Brindisi
- Department of Pharmacy, Department of Excellence 2018-2022, University of Naples Federico II, Naples, Italy
| |
Collapse
|
13
|
Girola N, Resende-Lara PT, Figueiredo CR, Massaoka MH, Azevedo RA, Cunha RLOR, Polonelli L, Travassos LR. Molecular, Biological and Structural Features of V L CDR-1 Rb44 Peptide, Which Targets the Microtubule Network in Melanoma Cells. Front Oncol 2019; 9:25. [PMID: 30740361 PMCID: PMC6355703 DOI: 10.3389/fonc.2019.00025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 01/08/2019] [Indexed: 12/21/2022] Open
Abstract
Microtubules are important drug targets in tumor cells, owing to their role in supporting and determining the cell shape, organelle movement and cell division. The complementarity-determining regions (CDRs) of immunoglobulins have been reported to be a source of anti-tumor peptide sequences, independently of the original antibody specificity for a given antigen. We found that, the anti-Lewis B mAb light-chain CDR1 synthetic peptide Rb44, interacted with microtubules and induced depolymerization, with subsequent degradation of actin filaments, leading to depolarization of mitochondrial membrane-potential, increase of ROS, cell cycle arrest at G2/M, cleavage of caspase-9, caspase-3 and PARP, upregulation of Bax and downregulation of Bcl-2, altogether resulting in intrinsic apoptosis of melanoma cells. The in vitro inhibition of angiogenesis was also an Rb44 effect. Peritumoral injection of Rb44L1 delayed growth of subcutaneously grafted melanoma cells in a syngeneic mouse model. L1-CDRs from immunoglobulins and their interactions with tubulin-dimers were explored to interpret effects on microtubule stability. The opening motion of tubulin monomers allowed for efficient L1-CDR docking, impairment of dimer formation and microtubule dissociation. We conclude that Rb44 VL-CDR1 is a novel peptide that acts on melanoma microtubule network causing cell apoptosis in vitro and melanoma growth inhibition in vivo.
Collapse
Affiliation(s)
- Natalia Girola
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit, Federal University of São Paulo, São Paulo, Brazil
| | - Pedro T Resende-Lara
- Computational Biology and Bioinformatics Laboratory, Federal University of ABC, Santo André, Brazil
| | - Carlos R Figueiredo
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit, Federal University of São Paulo, São Paulo, Brazil.,Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | | | - Ricardo A Azevedo
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit, Federal University of São Paulo, São Paulo, Brazil
| | - Rodrigo L O R Cunha
- Chemical Biology Laboratory, Natural and Human Sciences Center, Federal University of ABC, Santo André, Brazil
| | - Luciano Polonelli
- Unit of Biomedical, Biotechnological and Translational Sciences, Department of Medicine and Surgery, Universitá degli Studi di Parma, Parma, Italy
| | - Luiz R Travassos
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit, Federal University of São Paulo, São Paulo, Brazil.,Recepta Biopharma, São Paulo, Brazil
| |
Collapse
|
14
|
Brindisi M, Ulivieri C, Alfano G, Gemma S, de Asís Balaguer F, Khan T, Grillo A, Chemi G, Menchon G, Prota AE, Olieric N, Lucena-Agell D, Barasoain I, Diaz JF, Nebbioso A, Conte M, Lopresti L, Magnano S, Amet R, Kinsella P, Zisterer DM, Ibrahim O, O'Sullivan J, Morbidelli L, Spaccapelo R, Baldari C, Butini S, Novellino E, Campiani G, Altucci L, Steinmetz MO, Brogi S. Structure-activity relationships, biological evaluation and structural studies of novel pyrrolonaphthoxazepines as antitumor agents. Eur J Med Chem 2018; 162:290-320. [PMID: 30448418 DOI: 10.1016/j.ejmech.2018.11.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/11/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023]
Abstract
Microtubule-targeting agents (MTAs) are a class of clinically successful anti-cancer drugs. The emergence of multidrug resistance to MTAs imposes the need for developing new MTAs endowed with diverse mechanistic properties. Benzoxazepines were recently identified as a novel class of MTAs. These anticancer agents were thoroughly characterized for their antitumor activity, although, their exact mechanism of action remained elusive. Combining chemical, biochemical, cellular, bioinformatics and structural efforts we developed improved pyrrolonaphthoxazepines antitumor agents and their mode of action at the molecular level was elucidated. Compound 6j, one of the most potent analogues, was confirmed by X-ray as a colchicine-site MTA. A comprehensive structural investigation was performed for a complete elucidation of the structure-activity relationships. Selected pyrrolonaphthoxazepines were evaluated for their effects on cell cycle, apoptosis and differentiation in a variety of cancer cells, including multidrug resistant cell lines. Our results define compound 6j as a potentially useful optimized hit for the development of effective compounds for treating drug-resistant tumors.
Collapse
Affiliation(s)
- Margherita Brindisi
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, I-53100, Siena, Italy; Istituto Toscano Tumori, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy
| | - Cristina Ulivieri
- Istituto Toscano Tumori, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy; Department of Life Sciences, via Aldo Moro 2, I-53100, Siena, Italy
| | - Gloria Alfano
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, I-53100, Siena, Italy; Istituto Toscano Tumori, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy
| | - Sandra Gemma
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, I-53100, Siena, Italy; Istituto Toscano Tumori, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy
| | - Francisco de Asís Balaguer
- Department of Physical and Chemical Biology, Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Tuhina Khan
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, I-53100, Siena, Italy
| | - Alessandro Grillo
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, I-53100, Siena, Italy; Istituto Toscano Tumori, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy
| | - Giulia Chemi
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, I-53100, Siena, Italy; Istituto Toscano Tumori, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy
| | - Grégory Menchon
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland; University of Basel, Biozentrum, 4056 Basel, Switzerland
| | - Andrea E Prota
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland; University of Basel, Biozentrum, 4056 Basel, Switzerland
| | - Natacha Olieric
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland; University of Basel, Biozentrum, 4056 Basel, Switzerland
| | - Daniel Lucena-Agell
- Department of Physical and Chemical Biology, Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Isabel Barasoain
- Department of Physical and Chemical Biology, Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - J Fernando Diaz
- Department of Physical and Chemical Biology, Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Angela Nebbioso
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Vico L. de Crecchio 7, 80138, Naples, Italy
| | | | - Ludovica Lopresti
- Istituto Toscano Tumori, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy; Department of Life Sciences, via Aldo Moro 2, I-53100, Siena, Italy
| | - Stefania Magnano
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160, Pearse Street, Dublin 2, Ireland
| | - Rebecca Amet
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160, Pearse Street, Dublin 2, Ireland
| | - Paula Kinsella
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160, Pearse Street, Dublin 2, Ireland
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160, Pearse Street, Dublin 2, Ireland
| | - Ola Ibrahim
- School of Dental Science, Trinity College Dublin, Lincoln Place, Dublin 2, Ireland
| | - Jeff O'Sullivan
- School of Dental Science, Trinity College Dublin, Lincoln Place, Dublin 2, Ireland
| | - Lucia Morbidelli
- Istituto Toscano Tumori, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy; Department of Life Sciences, via Aldo Moro 2, I-53100, Siena, Italy
| | - Roberta Spaccapelo
- Department of Experimental Medicine, University of Perugia, P.le Gambuli, I-06132, Perugia, Italy
| | - Cosima Baldari
- Istituto Toscano Tumori, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy; Department of Life Sciences, via Aldo Moro 2, I-53100, Siena, Italy
| | - Stefania Butini
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, I-53100, Siena, Italy; Istituto Toscano Tumori, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy.
| | - Ettore Novellino
- Department of Pharmacy, University of Napoli Federico II, DoE Department of Excellence 2018-2022, Via D. Montesano 49, 80131, Napoli, Italy
| | - Giuseppe Campiani
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, I-53100, Siena, Italy; Istituto Toscano Tumori, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy.
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Vico L. de Crecchio 7, 80138, Naples, Italy
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland; University of Basel, Biozentrum, 4056 Basel, Switzerland
| | - Simone Brogi
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, I-53100, Siena, Italy; Istituto Toscano Tumori, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy; Department of Pharmacy, University of Napoli Federico II, DoE Department of Excellence 2018-2022, Via D. Montesano 49, 80131, Napoli, Italy
| |
Collapse
|
15
|
Brogi S, Maramai S, Brindisi M, Chemi G, Porcari V, Corallo C, Gennari L, Novellino E, Ramunno A, Butini S, Campiani G, Gemma S. Activation of the Wnt Pathway by Small Peptides: Rational Design, Synthesis and Biological Evaluation. ChemMedChem 2017; 12:2074-2085. [PMID: 29131552 DOI: 10.1002/cmdc.201700551] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/03/2017] [Indexed: 12/13/2022]
Abstract
A computational analysis of the X-ray structure of the low-density lipoprotein receptor-related protein 6 (LRP6) with the Dickkopf-1 (DKK1) C-terminal fragment has allowed us to rationally design a small set of decapeptides. These compounds behave as agonists of the canonical Wnt pathway in the micromolar range when tested on a dual luciferase Wnt functional assay in glioblastoma cells. Two of the oligopeptides showed a lack of cytotoxicity in human primary osteoblasts isolated from sponge bone tissue (femoral heads or knees of elderly patients). According to the mechanism of action, the studies revealed a dose- and time-dependent increase in the viability of human osteoblasts. These results may indicate a potential therapeutic application of this class of compounds in the treatment of bone diseases related to aging, such as osteoporosis.
Collapse
Affiliation(s)
- Simone Brogi
- European Research Centre for Drug Discovery and Development, NatSynDrugs and Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Samuele Maramai
- European Research Centre for Drug Discovery and Development, NatSynDrugs and Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Margherita Brindisi
- European Research Centre for Drug Discovery and Development, NatSynDrugs and Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Giulia Chemi
- European Research Centre for Drug Discovery and Development, NatSynDrugs and Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Valentina Porcari
- Siena Biotech S.p.A., Strada del Petriccio e Belriguardo 35, Siena, 53100, Italy
| | - Claudio Corallo
- Department of Medical, Surgical and Neurological Sciences, S. Maria alle Scotte Hospital Siena, University of Siena, viale Mario Bracci 1, 53100, Siena, Italy
| | - Luigi Gennari
- Department of Medical, Surgical and Neurological Sciences, S. Maria alle Scotte Hospital Siena, University of Siena, viale Mario Bracci 1, 53100, Siena, Italy
| | - Ettore Novellino
- Dipartimento di Farmacia, University of Naples Federico II, via D. Montesano 49, 80131, Naples, Italy
| | - Anna Ramunno
- Dipartimento di Farmacia/DIFARMA, University of Salerno, via Giovanni Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Stefania Butini
- European Research Centre for Drug Discovery and Development, NatSynDrugs and Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Giuseppe Campiani
- European Research Centre for Drug Discovery and Development, NatSynDrugs and Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Sandra Gemma
- European Research Centre for Drug Discovery and Development, NatSynDrugs and Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| |
Collapse
|
16
|
Wang X, Lin M, Xu D, Lai D, Zhou L. Structural Diversity and Biological Activities of Fungal Cyclic Peptides, Excluding Cyclodipeptides. Molecules 2017; 22:E2069. [PMID: 29186926 PMCID: PMC6150023 DOI: 10.3390/molecules22122069] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 11/20/2017] [Accepted: 11/23/2017] [Indexed: 11/23/2022] Open
Abstract
Cyclic peptides are cyclic compounds formed mainly by the amide bonds between either proteinogenic or non-proteinogenic amino acids. This review highlights the occurrence, structures and biological activities of fungal cyclic peptides (excluding cyclodipeptides, and peptides containing ester bonds in the core ring) reported until August 2017. About 293 cyclic peptides belonging to the groups of cyclic tri-, tetra-, penta-, hexa-, hepta-, octa-, nona-, deca-, undeca-, dodeca-, tetradeca-, and octadecapeptides as well as cyclic peptides containing ether bonds in the core ring have been isolated from fungi. They were mainly isolated from the genera Aspergillus, Penicillium, Fusarium, Acremonium and Amanita. Some of them were screened to have antimicrobial, antiviral, cytotoxic, phytotoxic, insecticidal, nematicidal, immunosuppressive and enzyme-inhibitory activities to show their potential applications. Some fungal cyclic peptides such as the echinocandins, pneumocandins and cyclosporin A have been developed as pharmaceuticals.
Collapse
Affiliation(s)
- Xiaohan Wang
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| | - Minyi Lin
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| | - Dan Xu
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| | - Daowan Lai
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| | - Ligang Zhou
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
17
|
First dual AK/GSK-3β inhibitors endowed with antioxidant properties as multifunctional, potential neuroprotective agents. Eur J Med Chem 2017; 138:438-457. [PMID: 28689095 DOI: 10.1016/j.ejmech.2017.06.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/07/2017] [Accepted: 06/08/2017] [Indexed: 02/02/2023]
Abstract
The manuscript deals with the design, synthesis and biological evaluation of novel benzoxazinone-based and indole-based compounds as multifunctional neuroprotective agents. These compounds inhibit human adenosine kinase (hAK) and human glycogen synthase kinase 3 beta (hGSK-3β) enzymes. Computational analysis based on a molecular docking approach underlined the potential structural requirements for simultaneously targeting both proteins' allosteric sites. In silico hints drove the synthesis of appropriately decorated benzoxazinones and indoles (5a-s, and 6a-c) and biochemical analysis revealed their behavior as allosteric inhibitors of hGSK-3β. For both our hit 4 and the best compounds of the series (5c,l and 6b) the potential antioxidant profile was assessed in human neuroblastoma cell lines (IMR 32, undifferentiated and neuronal differentiated), by evaluating the protective effect of selected compounds against H2O2 cytotoxicity and reactive oxygen species (ROS) production. Results showed a strong efficacy of the tested compounds, even at the lower doses, in counteracting the induced oxidative stress (50 μM of H2O2) and in preventing ROS formation. In addition, the tested compounds did not show any cytotoxic effect determined by the LDH release, at the concentration range analyzed (from 0.1 to 50 μM). This study allowed the identification of compound 5l, as the first dual hAK/hGSK-3β inhibitor reported to date. Compound 5l, which behaves as an effective antioxidant, holds promise for the development of new series of potential therapeutic agents for the treatment of neurodegenerative diseases characterized by an innovative pharmacological profile.
Collapse
|
18
|
Wu G, Kim D, Park BK, Park S, Ha JH, Kim TH, Gautam A, Kim JN, Lee SI, Park HB, Kim YS, Kwon HJ, Lee Y. Anti-metastatic effect of the TM4SF5-specific peptide vaccine and humanized monoclonal antibody on colon cancer in a mouse lung metastasis model. Oncotarget 2016; 7:79170-79186. [PMID: 27816969 PMCID: PMC5346706 DOI: 10.18632/oncotarget.13005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 10/19/2016] [Indexed: 12/17/2022] Open
Abstract
Transmembrane 4 superfamily member 5 protein (TM4SF5) is a potential therapeutic target for hepatocellular carcinoma (HCC) and colon cancer. In a previous study, we demonstrated the prophylactic and therapeutic effects of a TM4SF5-specific peptide vaccine and monoclonal antibody in HCC and colon cancer in a mouse model. Here, we designed a cyclic peptide targeting TM4SF5. Cyclic peptide-specific antibodies were produced in mice after immunization with a complex of the peptide, CpG-DNA, and liposomes. Intravenous injection of the CT-26 mouse colon cancer cell line into mice induced tumors in the lung. Immunization with the peptide vaccine improved the survival rate and reduced the growth of lung tumors. We established a monoclonal antibody specific to the cyclic TM4SF5-based peptide and humanized the antibody sequence by complementarity determining region-grafting. The humanized antibody was reactive to the cyclic peptide and TM4SF5 protein. Treatment of CT-26 cells with the humanized antibody reduced cell motility in vitro. Furthermore, direct injection of the humanized anti-TM4SF5 antibody in vivo reduced growth of lung tumors in mouse metastasis model. Therefore, we conclude that the immunization with the cyclic peptide vaccine and injection of the TM4SF5-specifc humanized antibody have an anti-metastatic effect against colon cancer in mice. Importantly, the humanized antibody may serve as a starting platf.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/pharmacology
- Cell Line, Tumor
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Colonic Neoplasms/drug therapy
- Colonic Neoplasms/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- HCT116 Cells
- Humans
- Injections, Intravenous
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/secondary
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Mice
- Peptides, Cyclic/administration & dosage
- Peptides, Cyclic/pharmacology
- Treatment Outcome
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Guang Wu
- Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Dongbum Kim
- Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Byoung Kwon Park
- Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Sangkyu Park
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Ji-Hee Ha
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Te Ha Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Avishekh Gautam
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jung Nam Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Su In Lee
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Han-Bum Park
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Yong-Sung Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hyung-Joo Kwon
- Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Younghee Lee
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| |
Collapse
|