251
|
Puerto Galvis CE, Kouznetsov VV. Synthesis of zanthoxylamide protoalkaloids and their in silico ADME-Tox screening and in vivo toxicity assessment in zebrafish embryos. Eur J Pharm Sci 2019; 127:291-299. [DOI: 10.1016/j.ejps.2018.10.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/25/2018] [Accepted: 10/30/2018] [Indexed: 01/23/2023]
|
252
|
Chemoinformatic Analysis of Selected Cacalolides from Psacalium decompositum (A. Gray) H. Rob. & Brettell and Psacalium peltatum (Kunth) Cass. and Their Effects on FcεRI-Dependent Degranulation in Mast Cells. Molecules 2018; 23:molecules23123367. [PMID: 30572603 PMCID: PMC6321304 DOI: 10.3390/molecules23123367] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/06/2018] [Accepted: 12/15/2018] [Indexed: 11/17/2022] Open
Abstract
Cacalolides are a kind of sesquiterpenoids natural compounds synthesized by Psacalium decompositum (A. Gray) H. Rob. & Brettell or Psacalium peltatum (Kunth) Cass. Antioxidant and hypoglycemic effects have been found for cacalolides such as cacalol, cacalone or maturine, however, their effects on inflammatory processes are still largely unclear. The main aim of this study was to investigate the biological activities of secondary metabolites from P. decompositum and P. peltatum through two approaches: (1) chemoinformatic and toxicoinformatic analysis based on ethnopharmacologic background; and (2) the evaluation of their potential anti-inflammatory/anti-allergic effects in bone marrow-derived mast cells by IgE/antigen complexes. The bioinformatics properties of the compounds: cacalol; cacalone; cacalol acetate and maturin acetate were evaluated through Osiris DataWarrior software and Molinspiration and PROTOX server. In vitro studies were performed to test the ability of these four compounds to inhibit antigen-dependent degranulation and intracellular calcium mobilization, as well as the production of reactive oxygen species in bone marrow-derived mast cells. Our findings showed that cacalol displayed better bioinformatics properties, also exhibited a potent inhibitory activity on IgE/antigen-dependent degranulation and significantly reduced the intracellular calcium mobilization on mast cells. These data suggested that cacalol could reduce the negative effects of the mast cell-dependent inflammatory process.
Collapse
|
253
|
Erukainure OL, Oyebode OA, Chukwuma CI, Matsabisa MG, Koorbanally NA, Islam MS. Raffia palm (Raphia hookeri) wine inhibits glucose diffusion; improves antioxidative activities; and modulates dysregulated pathways and metabolites in oxidative pancreatic injury. J Food Biochem 2018; 43:e12749. [PMID: 31353563 DOI: 10.1111/jfbc.12749] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/18/2018] [Accepted: 10/25/2018] [Indexed: 12/15/2022]
Abstract
Raffia palm wine is a natural drink from the stem of Raffia palm (Raphia hookeri) tree with nutritional and medicinal properties. The effect of fermentation was investigated on its antidiabetic and antioxidative effects in yeast cells and pancreatic tissues, respectively. Both unfermented and fermented palm wine significantly increased glucose uptake, reduced glutathione level (GSH), superoxide dismutase, and catalase activities. They also inhibited glucose diffusion, myeloperoxidase, and ATPase activities as well as decreased malondialdehyde and nitric oxide levels. They also led to the inactivation of oxidative metabolic pathways in oxidative pancreas with the generation of adenosine, sugar and inositol metabolites, selenium (enzyme co-factor) and vitamin metabolites owing to concomitant activation of vitamins, lipid, steroids, inositol, and sulfate/sulfite metabolic pathways. The results suggest the antidiabetic and antioxidative potentials of unfermented and fermented palm wine and may be attributed to the LC-MS-identified compounds which were mainly polyphenols and its glycosides, vitamins, and amino acids. PRACTICAL APPLICATIONS: Raffia palm wine is among the natural beverages employed for social, nutritional, and medicinal purposes. However, there are limited studies on its medicinal properties. This study reports for the first time, the ability of Raffia palm wine to stimulate glucose uptake, inhibit glucose diffusion, and ameliorate pancreatic oxidative injury, as well as the possible associated metabolic pathways that may be involved. These findings will further contribute in understanding the antidiabetic effect of Raffia palm wine, and the possible metabolic pathways involved.
Collapse
Affiliation(s)
- Ochuko L Erukainure
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal (Westville Campus), Durban, South Africa.,Nutrition and Toxicology Division, Federal Institute of Industrial Research, Lagos, Nigeria
| | - Olajumoke A Oyebode
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal (Westville Campus), Durban, South Africa
| | - Chika I Chukwuma
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal (Westville Campus), Durban, South Africa.,Department of Pharmacology, University of the Free State, Bloemfontein, South Africa
| | | | - Neil A Koorbanally
- School of Chemistry and Physics, University of KwaZulu-Natal (Westville Campus), Durban, South Africa
| | - Md Shahidul Islam
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal (Westville Campus), Durban, South Africa
| |
Collapse
|
254
|
Chemical Composition and Antifungal In Vitro and In Silico, Antioxidant, and Anticholinesterase Activities of Extracts and Constituents of Ouratea fieldingiana (DC.) Baill. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:1748487. [PMID: 30524481 PMCID: PMC6247570 DOI: 10.1155/2018/1748487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 08/02/2018] [Accepted: 08/12/2018] [Indexed: 12/25/2022]
Abstract
Ouratea fieldingiana (Gardner) Engl is popularly used for wound healing. This study describes the main chemical compounds present in extracts of O. fieldingiana and evaluates their biological potential by investigating antifungal, antioxidant, and anticholinesterase activities. The action mechanism of main antifungal compound was investigated by molecular docking using the enzyme sterol 14-α demethylase, CYP51, required for ergosterol biosynthesis. The seeds and leaves were extracted with ethanol in a Soxhlet apparatus and by maceration, respectively. Both extracts were subjected to silica gel column chromatography for isolation of main constituents, followed by purification in sephadex. The structures of compounds were established by 1H and 13C-NMR spectroscopy and identified by comparison with literature data as amentoflavone and kaempferol 3-O-rutinoside, respectively. The antioxidant activities of the extracts were determined by the DPPH and ABTS free radical inhibition methods. In general, the extracts with the highest antioxidant activity corresponded to those with higher content of phenolic compounds and flavonoids. The ethanol extracts and two isolated compounds presented relevant antifungal activity against several Candida strains. The in silico findings revealed that the compound amentoflavone coupled with the CYP450 protein due to the low energy stabilization (-9.39 kcal/mol), indicating a possible mechanism of action by inhibition of the ergosterol biosynthesis of Candida fungi.
Collapse
|
255
|
Kant K, Lal UR, Kumar A, Ghosh M. A merged molecular docking, ADME-T and dynamics approaches towards the genus of Arisaema as herpes simplex virus type 1 and type 2 inhibitors. Comput Biol Chem 2018; 78:217-226. [PMID: 30579134 DOI: 10.1016/j.compbiolchem.2018.12.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 01/01/2023]
Abstract
An attempt toward screening of phytoconstituents (Arisaema genus) against herpes viruses (HSV-1 and HSV-2) was carried out using in silico approaches. Human HSV-1 and HSV-2 are accountable for cold sores genital herpes, respectively. Two drug targets, namely thymidine kinase (TK; PDB: 2ki5) serine protease (PDB: 1at3) were selected for HSV-1 and HSV-2. Initially, molecular docking tool was employed to screened apex hits phytoconstituents against herpes infections. ADME-T studies of top ranked were also further highlighted to achieve their effectiveness. Following, molecular dynamics studies were also examined to further optimize the stability of ligands. Glide scores and binding interactions of phytoconstituents were compared with Acyclovir, the main drug used in treatment of HSV, the screened top hits exhibited more glide scores and better binding for both HSV-1 and HSV-2 receptors. Additionally, ADME-T showed an ideal range for top hits while molecular dynamics results also illustrated stability of models. Ultimately, the whole efforts reveal to top three most promising hits for HSV-1 (39, 21, 19) and HSV-2 (20, 51, 19) receptors which can be explored further in wet lab experiments as promising agents against HSV infections.
Collapse
Affiliation(s)
- Kamal Kant
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| | - Uma Ranjan Lal
- School of Pharmaceutical Sciences, Shoolini University, Post Box 9, Solan 173212, Himachal Pradesh, India
| | - Anoop Kumar
- Department of Pharmacology, Indo-Soviet Friendship College of Pharmacy (ISFCP), Ghal Kalan, Moga 142001, Punjab, India.
| | - Manik Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India.
| |
Collapse
|
256
|
Lysenko A, Sharma A, Boroevich KA, Tsunoda T. An integrative machine learning approach for prediction of toxicity-related drug safety. Life Sci Alliance 2018; 1:e201800098. [PMID: 30515477 PMCID: PMC6262234 DOI: 10.26508/lsa.201800098] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 11/20/2018] [Accepted: 11/20/2018] [Indexed: 01/28/2023] Open
Abstract
Recent trends in drug development have been marked by diminishing returns caused by the escalating costs and falling rates of new drug approval. Unacceptable drug toxicity is a substantial cause of drug failure during clinical trials and the leading cause of drug withdraws after release to the market. Computational methods capable of predicting these failures can reduce the waste of resources and time devoted to the investigation of compounds that ultimately fail. We propose an original machine learning method that leverages identity of drug targets and off-targets, functional impact score computed from Gene Ontology annotations, and biological network data to predict drug toxicity. We demonstrate that our method (TargeTox) can distinguish potentially idiosyncratically toxic drugs from safe drugs and is also suitable for speculative evaluation of different target sets to support the design of optimal low-toxicity combinations.
Collapse
Affiliation(s)
- Artem Lysenko
- Laboratory for Medical Science Mathematics, Rikagaku Kenkyūjyo Center for Integrative Medical Sciences, Tsurumi, Japan
| | - Alok Sharma
- Laboratory for Medical Science Mathematics, Rikagaku Kenkyūjyo Center for Integrative Medical Sciences, Tsurumi, Japan
- School of Engineering and Physics, University of the South Pacific, Suva, Fiji
| | - Keith A Boroevich
- Laboratory for Medical Science Mathematics, Rikagaku Kenkyūjyo Center for Integrative Medical Sciences, Tsurumi, Japan
| | - Tatsuhiko Tsunoda
- Laboratory for Medical Science Mathematics, Rikagaku Kenkyūjyo Center for Integrative Medical Sciences, Tsurumi, Japan
- Department of Medical Science Mathematics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
- Core Research for Evolutionary Science and Technology Program, Japan Science and Technology Agency, Tokyo, Japan
| |
Collapse
|
257
|
Olotu FA, Munsamy G, Soliman MES. Does Size Really Matter? Probing the Efficacy of Structural Reduction in the Optimization of Bioderived Compounds - A Computational "Proof-of-Concept". Comput Struct Biotechnol J 2018; 16:573-586. [PMID: 30546858 PMCID: PMC6280605 DOI: 10.1016/j.csbj.2018.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/14/2018] [Accepted: 11/18/2018] [Indexed: 02/07/2023] Open
Abstract
Over the years, numerous synthetic approaches have been utilized in drug design to improve the pharmacological properties of naturally derived compounds and most importantly, minimize toxic effects associated with their transition to drugs. The reduction of complex bioderived compounds to simpler bioactive fragments has been identified as a viable strategy to develop lead compounds with improved activities and minimal toxicities. Although this ‘reductive’ strategy has been widely exemplified, underlying biological events remain unresolved, hence the unanswered question remains how does the fragmentation of a natural compound improve its bioactivity and reduce toxicities? Herein, using a combinatorial approach, we initialize a computational “proof-of- concept” to expound the differential pharmacological and antagonistic activities of a natural compound, Anguinomycin D, and its synthetic fragment, SB640 towards Exportin Chromosome Region Maintenance 1 (CRM1). Interestingly, our findings revealed that in comparison with the parent compound, SB640 exhibited improved pharmacological attributes, while toxicities and off-target activities were relatively minimal. Moreover, we observed that the reduced size of SB640 allowed ‘deep access’ at the Nuclear Export Signals (NES) binding groove of CRM1, which favored optimal and proximal positioning towards crucial residues while the presence of the long polyketide tail in Anguinomycin D constrained its burial at the hydrophobic groove. Furthermore, with regards to their antagonistic functions, structural inactivation (rigidity) was more pronounced in CRM1 when bound by SB640 as compared to Anguinomycin D. These findings provide essential insights that portray synthetic fragmentation of natural compounds as a feasible approach towards the discovery of potential leads in disease treatment.
Collapse
Affiliation(s)
- Fisayo A Olotu
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| | - Geraldene Munsamy
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| |
Collapse
|
258
|
Rivera-Pérez WA, Yépes-Pérez AF, Martínez-Pabón MC. Molecular docking and in silico studies of the physicochemical properties of potential inhibitors for the phosphotransferase system of Streptococcus mutans. Arch Oral Biol 2018; 98:164-175. [PMID: 30500666 DOI: 10.1016/j.archoralbio.2018.09.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 10/27/2022]
Abstract
This study identified potential inhibitory compounds of the phosphoenolpyruvate-sugar. Phosphotransferase system of S. mutans, specifically enzyme II mannose transporter (EIIMan) in its subunits IIA, IIB and IIC by means of a selection protocol and in silico molecular analysis. Intervening the phosphotransferase system would compromise the physiological behavior and the pathogenic expression of S. mutans, and possibly other acidogenic bacteria that use phosphotransferases in their metabolism-making the phosphotransferase system a therapeutic target for the selective control of acidogenic microorganisms in caries control. Several computational techniques were used to evaluate molecular, physicochemical, and toxicological aspects of various compounds. Molecular docking was used to calculate the binding potential (ΔG) between receptor protein subunits and more than 836,000 different chemical compounds from the ZINC database. Physicochemical parameters related to the compounds' pharmacokinetic and pharmacodynamic indicators were evaluated, including absorption, distribution, metabolism, excretion, and toxicity (ADMET), and chemical analysis characterized the compounds structures. Thirteen compounds with EII binding potential of the phosphotransferase system of S. mutans and favorable ADMET properties were identified. Six spirooxindoles and three pyrrolidones stand out from the found compounds; unique structural characteristics of spirooxindoles and pyrrolidones associated with various reported biological activities like anti-microbial, antiinflammatory, anticancer, nootropic, neuroprotective and antiepileptic effects, among other pharmacological effects with surprising differences in terms of mechanisms of action. Following studies will provide more evidence of the action of these compounds on the phosphotransferase system of S. mutans, and its possible applications.
Collapse
Affiliation(s)
- Wbeimar Andrey Rivera-Pérez
- Faculty of Dentistry, University of Antioquia- UdeA, 64 Street No. 52-59, Block 31, Oral Microbiology Laboratory No. 216, Health Area, Medellin, Colombia.
| | - Andrés Felipe Yépes-Pérez
- Exact and Natural Sciences School, University of Antioquia-UdeA, Universidad de Antioquia. 67 street No. 53-108, Block 2, Chemistry of Colombian, Plants Laboratory, Office 330, Medellin, Colombia.
| | - Maria Cecilia Martínez-Pabón
- Faculty of Dentistry, University of Antioquia- UdeA, 64 Street No. 52-59, Block 31, Oral Microbiology Laboratory No. 216, Health Area, Medellin, Colombia.
| |
Collapse
|
259
|
Erukainure OL, Mopuri R, Chukwuma CI, Koorbanally NA, Islam MS. Phaseolus lunatus
(lima beans) abates Fe
2+
‐induced hepatic redox imbalance; inhibits intestinal glucose absorption and major carbohydrate catabolic enzymes; and modulates muscle glucose uptake. J Food Biochem 2018. [DOI: 10.1111/jfbc.12655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Ochuko L. Erukainure
- Department of Biochemistry, School of Life Sciences University of KwaZulu‐Natal (Westville Campus) Durban South Africa
- Nutrition and Toxicology Division Federal Institute of Industrial Research Lagos Nigeria
| | - Ramgopal Mopuri
- Department of Biochemistry, School of Life Sciences University of KwaZulu‐Natal (Westville Campus) Durban South Africa
| | - Chika I. Chukwuma
- Department of Biochemistry, School of Life Sciences University of KwaZulu‐Natal (Westville Campus) Durban South Africa
- Faculty of Health Sciences, Department of Pharmacology University of Free State Bloemfontein South Africa
| | - Neil A. Koorbanally
- School of Chemistry and Physics University of KwaZulu‐Natal (Westville Campus) Durban South Africa
| | - Md. Shahidul Islam
- Department of Biochemistry, School of Life Sciences University of KwaZulu‐Natal (Westville Campus) Durban South Africa
| |
Collapse
|
260
|
Olotu F, Adeniji E, Agoni C, Bjij I, Khan S, Elrashedy A, Soliman M. An update on the discovery and development of selective heat shock protein inhibitors as anti-cancer therapy. Expert Opin Drug Discov 2018; 13:903-918. [PMID: 30207185 DOI: 10.1080/17460441.2018.1516035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Over the years, not a single HSP inhibitor has progressed into the post-market phase of drug development despite the success recorded in various pre-clinical and clinical studies. The inability of existing drugs to specifically target oncogenic HSPs has majorly accounted for these setbacks. Recent combinatorial strategies that incorporated computer-aided drug design (CADD) techniques are geared towards the development of highly specific HSP inhibitors with increased activities and minimal toxicities. Areas covered: In this review, strategic therapeutic approaches that have recently aided the development of selective HSP inhibitors were highlighted. Also, the significant contributions of CADD techniques over the years were discussed in detail. This article further describes promising computational paradigms and their applications towards the discovery of highly specific inhibitors of oncogenic HSPs. Expert opinion: The recent shift towards highly selective and specific HSP inhibition has shown great promise as evidenced by the development of paralog/isoform-selective HSP drugs. It could be further augmented with computer-aided drug design strategies, which incorporate reliable methods that would greatly enhance the design and optimization of novel inhibitors with improved activities and minimal toxicities.
Collapse
Affiliation(s)
- Fisayo Olotu
- a Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences , University of KwaZulu-Natal , Durban , South Africa
| | - Emmanuel Adeniji
- a Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences , University of KwaZulu-Natal , Durban , South Africa
| | - Clement Agoni
- a Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences , University of KwaZulu-Natal , Durban , South Africa
| | - Imane Bjij
- a Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences , University of KwaZulu-Natal , Durban , South Africa
| | - Shama Khan
- a Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences , University of KwaZulu-Natal , Durban , South Africa
| | | | - Mahmoud Soliman
- a Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences , University of KwaZulu-Natal , Durban , South Africa
| |
Collapse
|
261
|
Richardson BG, Jain AD, Potteti HR, Lazzara PR, David BP, Tamatam CR, Choma E, Skowron K, Dye K, Siddiqui Z, Wang YT, Krunic A, Reddy SP, Moore TW. Replacement of a Naphthalene Scaffold in Kelch-like ECH-Associated Protein 1 (KEAP1)/Nuclear Factor (Erythroid-derived 2)-like 2 (NRF2) Inhibitors. J Med Chem 2018; 61:8029-8047. [PMID: 30122040 DOI: 10.1021/acs.jmedchem.8b01133] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Activators of nuclear factor-erythroid 2-related factor 2 (NRF2) could lead to promising therapeutics for prevention and treatment of oxidative stress and inflammatory disorders. Ubiquitination and subsequent degradation of the transcription factor NRF2 is mediated by Kelch-like ECH-associated protein-1 (KEAP1). Inhibition of the KEAP1/NRF2 interaction with small molecules leads to NRF2 activation. Previously, we and others described naphthalene-based NRF2 activators, but the 1,4-diaminonaphthalene scaffold may not represent a drug-like scaffold. Paying particular attention to aqueous solubility, metabolic stability, potency, and mutagenicity, we modified a previously known, naphthalene-based nonelectrophilic NRF2 activator to give a series of non-naphthalene and heterocyclic scaffolds. We found that, compared to previously reported naphthalene-based compounds, a 1,4-isoquinoline scaffold provides a better mutagenic profile without sacrificing potency, stability, or solubility.
Collapse
Affiliation(s)
- Benjamin G Richardson
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy , ‡Department of Pediatrics, College of Medicine , §UICentre for Drug Discovery , ⊥Mass Spectrometry Core at Research Resources Center , #University of Illinois Cancer Center , University of Illinois at Chicago , 833 South Wood Street , Chicago , Illinois 60612 , United States
| | - Atul D Jain
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy , ‡Department of Pediatrics, College of Medicine , §UICentre for Drug Discovery , ⊥Mass Spectrometry Core at Research Resources Center , #University of Illinois Cancer Center , University of Illinois at Chicago , 833 South Wood Street , Chicago , Illinois 60612 , United States
| | | | - Phillip R Lazzara
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy , ‡Department of Pediatrics, College of Medicine , §UICentre for Drug Discovery , ⊥Mass Spectrometry Core at Research Resources Center , #University of Illinois Cancer Center , University of Illinois at Chicago , 833 South Wood Street , Chicago , Illinois 60612 , United States
| | - Brian P David
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy , ‡Department of Pediatrics, College of Medicine , §UICentre for Drug Discovery , ⊥Mass Spectrometry Core at Research Resources Center , #University of Illinois Cancer Center , University of Illinois at Chicago , 833 South Wood Street , Chicago , Illinois 60612 , United States
| | | | - Ewelina Choma
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy , ‡Department of Pediatrics, College of Medicine , §UICentre for Drug Discovery , ⊥Mass Spectrometry Core at Research Resources Center , #University of Illinois Cancer Center , University of Illinois at Chicago , 833 South Wood Street , Chicago , Illinois 60612 , United States
| | - Kornelia Skowron
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy , ‡Department of Pediatrics, College of Medicine , §UICentre for Drug Discovery , ⊥Mass Spectrometry Core at Research Resources Center , #University of Illinois Cancer Center , University of Illinois at Chicago , 833 South Wood Street , Chicago , Illinois 60612 , United States
| | | | - Zamia Siddiqui
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy , ‡Department of Pediatrics, College of Medicine , §UICentre for Drug Discovery , ⊥Mass Spectrometry Core at Research Resources Center , #University of Illinois Cancer Center , University of Illinois at Chicago , 833 South Wood Street , Chicago , Illinois 60612 , United States
| | | | - Aleksej Krunic
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy , ‡Department of Pediatrics, College of Medicine , §UICentre for Drug Discovery , ⊥Mass Spectrometry Core at Research Resources Center , #University of Illinois Cancer Center , University of Illinois at Chicago , 833 South Wood Street , Chicago , Illinois 60612 , United States
| | | | - Terry W Moore
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy , ‡Department of Pediatrics, College of Medicine , §UICentre for Drug Discovery , ⊥Mass Spectrometry Core at Research Resources Center , #University of Illinois Cancer Center , University of Illinois at Chicago , 833 South Wood Street , Chicago , Illinois 60612 , United States
| |
Collapse
|
262
|
Günthardt BF, Hollender J, Hungerbühler K, Scheringer M, Bucheli TD. Comprehensive Toxic Plants-Phytotoxins Database and Its Application in Assessing Aquatic Micropollution Potential. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:7577-7588. [PMID: 29944838 DOI: 10.1021/acs.jafc.8b01639] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The production of toxic plant secondary metabolites (phytotoxins) for defense is a widespread phenomenon in the plant kingdom and is even present in agricultural crops. These phytotoxins may have similar characteristics to anthropogenic micropollutants in terms of persistence and toxicity. However, they are only rarely included in environmental risk assessments, partly because a systematic overview of phytotoxins is missing. Here, we present a newly developed, freely available database, Toxic Plants-PhytoToxins (TPPT), containing 1586 phytotoxins of potential ecotoxicological relevance in Central Europe linked to 844 plant species. Our database summarizes phytotoxin patterns in plant species and provides detailed biological and chemical information as well as in silico estimated properties. Using the database, we evaluated phytotoxins regarding occurrence, approximated from the frequencies of Swiss plant species; environmental behavior based on aquatic persistence and mobility; and toxicity. The assessment showed that over 34% of all phytotoxins are potential aquatic micropollutants and should be included in environmental investigations.
Collapse
Affiliation(s)
- Barbara F Günthardt
- Environmental Analytics , Agroscope , Reckenholzstrasse 191 , 8046 Zürich , Switzerland
- Institute of Biogeochemistry and Pollutant Dynamics , ETH Zurich , Universitätsstrasse 16 , 8092 Zürich , Switzerland
| | - Juliane Hollender
- Institute of Biogeochemistry and Pollutant Dynamics , ETH Zurich , Universitätsstrasse 16 , 8092 Zürich , Switzerland
- Swiss Federal Institute of Aquatic Science and Technology (Eawag) , Überlandstrasse 133 , 8600 Dübendorf , Switzerland
| | - Konrad Hungerbühler
- Institute for Chemical and Bioengineering , ETH Zurich , Wolfgang-Pauli-Strasse 10 , 8093 Zürich , Switzerland
| | - Martin Scheringer
- Institute for Chemical and Bioengineering , ETH Zurich , Wolfgang-Pauli-Strasse 10 , 8093 Zürich , Switzerland
- RECETOX , Masaryk University , Kamenice 753/5 , 625 00 Brno , Czech Republic
| | - Thomas D Bucheli
- Environmental Analytics , Agroscope , Reckenholzstrasse 191 , 8046 Zürich , Switzerland
| |
Collapse
|
263
|
A novel calix[4]pyrrole derivative as a potential anticancer agent that forms genotoxic adducts with DNA. Sci Rep 2018; 8:11075. [PMID: 30038406 PMCID: PMC6056420 DOI: 10.1038/s41598-018-29314-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 07/09/2018] [Indexed: 01/03/2023] Open
Abstract
meso-(p-acetamidophenyl)-calix[4]pyrrole 3 was found to exhibit remarkable cytotoxicity towards A549 cancer cells. A comparative study including the isomer of 3 meso-(m-acetamidophenyl)-calix[4]pyrrole 5, as well as molecules containing 'fragments' of these structures, demonstrated that both the calix[4]pyrrole and the acetamidophenyl units are essential for high cytotoxicity. Although calix[4]pyrroles and other anion-complexing ionophores have recently been reported to induce apoptosis by perturbing cellular chloride concentrations, in our study an alternative mechanism has emerged, as proven by the isolation of covalent DNA adducts revealed by the 32P postlabelling technique. Preliminary pharmacokinetic studies indicate that 3 is able to cross the Blood-Brain-Barrier, therefore being a potential drug that could kill primary and brain metastatic cancer cells simultaneously.
Collapse
|
264
|
Erukainure OL, Narainpersad N, Singh M, Olakunle S, Islam MS. Clerodendrum volubile inhibits key enzymes linked to type 2 diabetes but induces cytotoxicity in human embryonic kidney (HEK293) cells via exacerbated oxidative stress and proinflammation. Biomed Pharmacother 2018; 106:1144-1152. [PMID: 30119181 DOI: 10.1016/j.biopha.2018.07.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/29/2018] [Accepted: 07/01/2018] [Indexed: 12/12/2022] Open
Abstract
The toxicity and safety associated with the use of medicinal plants remains a major concern. In this study, the antidiabetic properties of the dichloromethane (DCM) fraction of C. volubile leaves were investigated in vitro. Its cytotoxic effect and mechanism of toxicity were also investigated in Human Embryonic Kidney (HEK293) cells. The fraction was subjected to in vitro antioxidant assays using the 2,2'-diphenyl-1-picrylhydrazyl (DPPH) scavenging and Ferric reducing antioxidant power (FRAP) protocols. Its enzyme-inhibitory properties were investigated on α-glucosidase and α-amylase activities. Gas Chromatography Mass Spectroscopy (GCMS) and Fourier Transform Infrared (FTIR) spectroscopic analysis were used to identify its phytoconstituents. Cytotoxicity was determined via MTT assay. The treated cells were assayed for reduced glutathione (GSH), non-protein thiol, nitric oxide and malondialdehyde (MDA) levels, as well as Superoxide Dismutase (SOD), catalase, myeloperoxidase and ATPase activities. Cell apoptosis and/or morphological changes were determined using the acridine orange and ethidium bromide (AO/EB) dual staining method. The fraction showed significant (p < 0.05) antioxidant and enzyme-inhibitory activity. It showed significant (p < 0.05) cytotoxic effect against HEK293 cells with concomitant depletion of antioxidative and elevation of proinflammatory biomarkers. Morphological changes were examined in the cells with an apoptotic index of 0.84. 1,1-Dodecanediol, diacetate was identified as the most predominant compound, while aromatics and amines as the most functional groups present in the fraction. These results suggest the antidiabetic and cytotoxic effects of C. volubile leaves. The toxicity can be attributed to induced oxidative stress and proinflammation with concomitant depletion of ATP leading to apoptosis of the cells.
Collapse
Affiliation(s)
- Ochuko L Erukainure
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa; Nutrition and Toxicology Division, Federal Institute of Industrial Research, Lagos, Nigeria
| | - Nicholisha Narainpersad
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Moganavelli Singh
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Sanni Olakunle
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Md Shahidul Islam
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa.
| |
Collapse
|
265
|
Looking for Novel Capsid Protein Multimerization Inhibitors of Feline Immunodeficiency Virus. Pharmaceuticals (Basel) 2018; 11:ph11030067. [PMID: 29996481 PMCID: PMC6161179 DOI: 10.3390/ph11030067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/02/2018] [Accepted: 07/04/2018] [Indexed: 12/25/2022] Open
Abstract
Feline immunodeficiency virus (FIV) is a member of the retroviridae family of viruses. It causes acquired immunodeficiency syndrome (AIDS) in worldwide domestic and non-domestic cats and is a cause of an important veterinary issue. The genome organization of FIV and the clinical characteristics of the disease caused by FIV are similar to human immunodeficiency virus (HIV). Both viruses infect T lymphocytes, monocytes, and macrophages, with a similar replication cycle in infected cells. Thus, the infection of cats with FIV is also a useful tool for the study and development of novel drugs and vaccines against HIV. Anti-retroviral drugs studied extensively with regards to HIV infection have targeted different steps of the virus replication cycle: (1) disruption of the interaction with host cell surface receptors and co-receptors; (2) inhibition of fusion of the virus and cell membranes; (3) blocking of the reverse transcription of viral genomic RNA; (4) interruption of nuclear translocation and integration of viral DNA into host genomes; (5) prevention of viral transcript processing and nuclear export; and (6) inhibition of virion assembly and maturation. Despite the great success of anti-retroviral therapy in slowing HIV progression in humans, a similar therapy has not been thoroughly investigated for FIV infection in cats, mostly because of the little structural information available for FIV proteins. The FIV capsid protein (CA) drives the assembly of the viral particle, which is a critical step in the viral replication cycle. During this step, the CA protein oligomerizes to form a protective coat that surrounds the viral genome. In this work, we perform a large-scale screening of four hundred molecules from our in-house library using an in vitro assembly assay of p24, combined with microscale thermophoresis, to estimate binding affinity. This screening led to the discovery of around four novel hits that inhibited capsid assembly in vitro. These may provide new antiviral drugs against FIV.
Collapse
|
266
|
Banerjee P, Eckert AO, Schrey AK, Preissner R. ProTox-II: a webserver for the prediction of toxicity of chemicals. Nucleic Acids Res 2018. [PMID: 29718510 DOI: 10.1093/nar/gky318%jnucleicacidsresearch] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
Abstract
Advancement in the field of computational research has made it possible for the in silico methods to offer significant benefits to both regulatory needs and requirements for risk assessments, and pharmaceutical industry to assess the safety profile of a chemical. Here, we present ProTox-II that incorporates molecular similarity, pharmacophores, fragment propensities and machine-learning models for the prediction of various toxicity endpoints; such as acute toxicity, hepatotoxicity, cytotoxicity, carcinogenicity, mutagenicity, immunotoxicity, adverse outcomes pathways (Tox21) and toxicity targets. The predictive models are built on data from both in vitro assays (e.g. Tox21 assays, Ames bacterial mutation assays, hepG2 cytotoxicity assays, Immunotoxicity assays) and in vivo cases (e.g. carcinogenicity, hepatotoxicity). The models have been validated on independent external sets and have shown strong performance. ProTox-II provides a freely available webserver for in silico toxicity prediction for toxicologists, regulatory agencies, computational and medicinal chemists, and all users without login at http://tox.charite.de/protox_II. The webserver takes a two-dimensional chemical structure as an input and reports the possible toxicity profile of the chemical for 33 models with confidence scores, and an overall toxicity radar chart along with three most similar compounds with known acute toxicity.
Collapse
Affiliation(s)
- Priyanka Banerjee
- Structural Bioinformatics Group, Institute for Physiology & ECRC, Charité - University Medicine Berlin, 10115 Berlin, Germany
| | - Andreas O Eckert
- Structural Bioinformatics Group, Institute for Physiology & ECRC, Charité - University Medicine Berlin, 10115 Berlin, Germany
| | - Anna K Schrey
- Structural Bioinformatics Group, Institute for Physiology & ECRC, Charité - University Medicine Berlin, 10115 Berlin, Germany
| | - Robert Preissner
- Structural Bioinformatics Group, Institute for Physiology & ECRC, Charité - University Medicine Berlin, 10115 Berlin, Germany.,BB3R - Berlin Brandenburg 3R Graduate School, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
267
|
Hieu DT, Anh DT, Hai PT, Huong LTT, Park EJ, Choi JE, Kang JS, Dung PTP, Han SB, Nam NH. Quinazoline-Based Hydroxamic Acids: Design, Synthesis, and Evaluation of Histone Deacetylase Inhibitory Effects and Cytotoxicity. Chem Biodivers 2018; 15:e1800027. [DOI: 10.1002/cbdv.201800027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/12/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Doan Thanh Hieu
- Hanoi University of Pharmacy; 13-15 Le Thanh Tong Hanoi Vietnam
| | - Duong Tien Anh
- Hanoi University of Pharmacy; 13-15 Le Thanh Tong Hanoi Vietnam
| | - Pham-The Hai
- Hanoi University of Pharmacy; 13-15 Le Thanh Tong Hanoi Vietnam
| | - Le-Thi-Thu Huong
- School of Medicine and Pharmacy; Vietnam National University; 144 XuanThuy Hanoi Vietnam
| | - Eun Jae Park
- College of Pharmacy; Chungbuk National University; 194-31, Osongsaengmyung-1, Heungdeok Cheongju Chungbuk 28160 Korea
| | - Jeong Eun Choi
- College of Pharmacy; Chungbuk National University; 194-31, Osongsaengmyung-1, Heungdeok Cheongju Chungbuk 28160 Korea
| | - Jong Soon Kang
- Bio-Evaluation Center; Korea Research Institute of Bioscience and Biotechnology; 30 Yeongudanji-ro, Ochang-eup, Chenogwon-gu Cheongju-si Chungcheongbuk-do 28116 Korea
| | | | - Sang-Bae Han
- College of Pharmacy; Chungbuk National University; 194-31, Osongsaengmyung-1, Heungdeok Cheongju Chungbuk 28160 Korea
| | - Nguyen-Hai Nam
- Hanoi University of Pharmacy; 13-15 Le Thanh Tong Hanoi Vietnam
| |
Collapse
|
268
|
Pathak RK, Gupta A, Shukla R, Baunthiyal M. Identification of new drug-like compounds from millets as Xanthine oxidoreductase inhibitors for treatment of Hyperuricemia: A molecular docking and simulation study. Comput Biol Chem 2018; 76:32-41. [PMID: 29906649 DOI: 10.1016/j.compbiolchem.2018.05.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/11/2018] [Accepted: 05/13/2018] [Indexed: 12/30/2022]
Abstract
Xanthine oxidoreductase plays an important role in formation of uric acid and its regulation during purine catabolism. Uncontrolled expression of this enzyme is responsible for overproduction and deposition of uric acid in blood that is potentially injurious because it can breakdown DNA and protein molecules, triggering many diseases. Human Xanthine oxidoreductase (HsXOR) is considered to be a pharmacological target for the treatment of hyperuricemia. Many of the HsXOR-inhibitor drugs such as Febuxostat and Allopurinol are known to have significant adverse effects. Therefore, there is an urgent need to develop new HsXOR-inhibitor drugs with less or no toxicity for the long-term treatment or prevention of hyperuricemia-related diseases. Many nutritious and medical functions have been reported in millets. Present work deals with identification of millet derived compounds in terms of their interaction with target, HsXOR through molecular docking and dynamic simulation studies. Of thirty two chosen compounds, Luteolin and Quercitin showed more binding affinity with HsXOR than reference drugs, Febuxostat and Allopurinol. Molecular dynamics simulations (20 ns long) revealed that Luteolin-protein complex was energetically more stable than Quercitin-protein complex. The millet derived compounds i.e. Luteolin and Quercitin showed binding energy -9.7 kcal/mol whereas the known drugs i.e. Febuxostat and Allopurinol showed binding energy -8.0 kcal/mol and -5.5 kcal/mol respectively. Based on the study, Luteolin possess high potential to be considered for trial as an inhibitor of HsXOR as it may regulate the pathway by inhibiting HsXOR. Further investigations are proposed to consider Luteolin for developing future drugs from millets and other natural sources.
Collapse
Affiliation(s)
- Rajesh Kumar Pathak
- Department of Biotechnology, Govind Ballabh Pant Institute of Engineering & Technology, Pauri Garhwal 246194, Uttarakhand, India
| | - Ayushi Gupta
- Department of Biotechnology, Govind Ballabh Pant Institute of Engineering & Technology, Pauri Garhwal 246194, Uttarakhand, India
| | - Rohit Shukla
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North Eastern Hill University, Shillong 793022, Meghalaya, India
| | - Mamta Baunthiyal
- Department of Biotechnology, Govind Ballabh Pant Institute of Engineering & Technology, Pauri Garhwal 246194, Uttarakhand, India.
| |
Collapse
|
269
|
Lawal M, Olotu FA, Soliman MES. Across the blood-brain barrier: Neurotherapeutic screening and characterization of naringenin as a novel CRMP-2 inhibitor in the treatment of Alzheimer's disease using bioinformatics and computational tools. Comput Biol Med 2018; 98:168-177. [PMID: 29860210 DOI: 10.1016/j.compbiomed.2018.05.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/08/2018] [Accepted: 05/08/2018] [Indexed: 11/16/2022]
Abstract
The discovery and developmental processes of CNS drugs have been limited by the inability of potential drug molecules to pass through the blood-brain barrier (BBB). This presents a significant setback in the treatment of neurodegenerative disorders such as Alzheimer's disease (AD), hence the need for compounds that can adhere strictly to the selective criteria of suitable CNS drugs. Collapsin response mediator protein-2 (CRMP-2) has been recently identified as a viable target in neurotherapeutics due to its involvement in the etiology of AD. As shown in previous studies, Naringenin (NAR), a small molecule derivative of Drynaria rhizome (DR) extract, specifically binds CRMP-2 and reduces its phosphorylation. This was shown to facilitate axonal regrowth, with improvement in cognition and learning. Herein, we report the first account of the use of cheminformatics techniques to define the CNS drug-suitability of NAR using selective criteria, coupled with the prediction of possible biological activities and toxicities. Also, we evaluated the mechanistic activity of NAR by modeling its molecular interaction with human CRMP-2 (hCRMP-2). Physicochemical analyses revealed the suitability of NAR as a CNS drug and its ability to transverse the BBB. Possible neurogenic, anti-carcinogenic and cardioprotective activities were also predicted. NAR exhibited favorable binding to CRMP-2 and formed strong bonds with active site residues, which accounts for its stabilization and affinity. Moreover, NAR induced notable conformational changes in CRMP-2, an occurrence that could possibly disrupt kinase-mediated phosphorylation. These findings will aid in the optimization of NAR and improve its neurotherapeutic activities in the treatment of AD.
Collapse
Affiliation(s)
- Maryam Lawal
- Molecular Modelling and Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal, Westville, Durban, 4000, South Africa
| | - Fisayo A Olotu
- Molecular Modelling and Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal, Westville, Durban, 4000, South Africa
| | - Mahmoud E S Soliman
- Molecular Modelling and Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal, Westville, Durban, 4000, South Africa.
| |
Collapse
|
270
|
Muhammad A, Arthur DE, Babangida S, Erukainure OL, Malami I, Sani H, Abdulhamid AW, Ajiboye IO, Saka AA, Hamza NM, Asema S, Ado ZM, Musa TI. Modulatory role of rutin on 2,5-hexanedione-induced chromosomal and DNA damage in rats: validation of computational predictions. Drug Chem Toxicol 2018; 43:113-126. [PMID: 29745774 DOI: 10.1080/01480545.2018.1465948] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The aim of this study was to evaluate the potentials of rutin on 2,5-hexanedione-induced toxicities. Two successive phases were involved using in silico and in vivo approaches. The in silico was adopted for potential oral toxicity and docking. The in vivo was carried-out in two stages for two weeks; the ameliorative (stage 1, first week), preventive, and curative studies (stage 2, extended to second week). In stage 1, rats were divided into four groups of seven each (distilled water, 3% (v/v) 2,5-hexanedione, 10 mg/kg rutin, and co-administration). In stage 2, the experimental groups were given either rutin or 2,5-hexanedione and treated in reverse order. Lipid peroxidation, protein carbonyl, and DNA fragmentation in tissues and bone marrow cells micronucleus were determined. The predicted Median lethal dose (LD50) of >5000 mg/kg and toxicity class of five (5) indicates the safety of rutin when orally administered. 2,5-Hexanedione comfortably docked in to the active sites of SOD (-22.857Kcal/mol; KI = 0.9621 µM), GPx (-11.2032Kcal/mol; KI = 0.9813 µM), and CAT (-16.446Kcal/mol; KI = 0.9726 µM) with strong hydrogen bond and hydrophobic interactions. However, only strong hydrophobic interaction was observed in the case of DNA (-3.3296Kcal/mol; KI = 0.9944). In vivo findings revealed deleterious effects of 2,5-hexanedione through induction of oxidative and chromosomal/DNA damage characterized by higher level of malondialdehyde, micronuclei formations, and DNA fragmentation. These have invariably, validates the findings from in silico experiments. Furthermore, rutin was able to ameliorate, protect, and reverse these effects, and was relatively non-toxic corroborating toxicity predictions. Rutin exhibited counteractive effects on 2,5-hexanedione-induced oxidative, chromosomal, and DNA damage.
Collapse
Affiliation(s)
- Aliyu Muhammad
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | | | - Sanusi Babangida
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | - Ochuko L Erukainure
- Nutrition and Toxicology Division, Federal Institute of Industrial Research, Oshodi, Nigeria
| | - Ibrahim Malami
- Department of Pharmacognosy and Ethnopharmacy, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Hadiza Sani
- Department of Medicine, Kaduna State University, Kaduna, Nigeria
| | | | | | - Ahmed Ariyo Saka
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | | | - Suleiman Asema
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | | | | |
Collapse
|
271
|
Banerjee P, Preissner R. BitterSweetForest: A Random Forest Based Binary Classifier to Predict Bitterness and Sweetness of Chemical Compounds. Front Chem 2018; 6:93. [PMID: 29696137 PMCID: PMC5905275 DOI: 10.3389/fchem.2018.00093] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/14/2018] [Indexed: 11/25/2022] Open
Abstract
Taste of a chemical compound present in food stimulates us to take in nutrients and avoid poisons. However, the perception of taste greatly depends on the genetic as well as evolutionary perspectives. The aim of this work was the development and validation of a machine learning model based on molecular fingerprints to discriminate between sweet and bitter taste of molecules. BitterSweetForest is the first open access model based on KNIME workflow that provides platform for prediction of bitter and sweet taste of chemical compounds using molecular fingerprints and Random Forest based classifier. The constructed model yielded an accuracy of 95% and an AUC of 0.98 in cross-validation. In independent test set, BitterSweetForest achieved an accuracy of 96% and an AUC of 0.98 for bitter and sweet taste prediction. The constructed model was further applied to predict the bitter and sweet taste of natural compounds, approved drugs as well as on an acute toxicity compound data set. BitterSweetForest suggests 70% of the natural product space, as bitter and 10% of the natural product space as sweet with confidence score of 0.60 and above. 77% of the approved drug set was predicted as bitter and 2% as sweet with a confidence score of 0.75 and above. Similarly, 75% of the total compounds from acute oral toxicity class were predicted only as bitter with a minimum confidence score of 0.75, revealing toxic compounds are mostly bitter. Furthermore, we applied a Bayesian based feature analysis method to discriminate the most occurring chemical features between sweet and bitter compounds using the feature space of a circular fingerprint.
Collapse
Affiliation(s)
| | - Robert Preissner
- Structural Bioinformatics Group, Institute for Physiology and ECRC, Charité – University Medicine Berlin, Berlin, Germany
| |
Collapse
|
272
|
Jain S, Kotsampasakou E, Ecker GF. Comparing the performance of meta-classifiers-a case study on selected imbalanced data sets relevant for prediction of liver toxicity. J Comput Aided Mol Des 2018; 32:583-590. [PMID: 29626291 PMCID: PMC5919997 DOI: 10.1007/s10822-018-0116-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 03/29/2018] [Indexed: 12/28/2022]
Abstract
Abstract Cheminformatics datasets used in classification problems, especially those related to biological or physicochemical properties, are often imbalanced. This presents a major challenge in development of in silico prediction models, as the traditional machine learning algorithms are known to work best on balanced datasets. The class imbalance introduces a bias in the performance of these algorithms due to their preference towards the majority class. Here, we present a comparison of the performance of seven different meta-classifiers for their ability to handle imbalanced datasets, whereby Random Forest is used as base-classifier. Four different datasets that are directly (cholestasis) or indirectly (via inhibition of organic anion transporting polypeptide 1B1 and 1B3) related to liver toxicity were chosen for this purpose. The imbalance ratio in these datasets ranges between 4:1 and 20:1 for negative and positive classes, respectively. Three different sets of molecular descriptors for model development were used, and their performance was assessed in 10-fold cross-validation and on an independent validation set. Stratified bagging, MetaCost and CostSensitiveClassifier were found to be the best performing among all the methods. While MetaCost and CostSensitiveClassifier provided better sensitivity values, Stratified Bagging resulted in high balanced accuracies. Graphical Abstract ![]()
Electronic supplementary material The online version of this article (10.1007/s10822-018-0116-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sankalp Jain
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Eleni Kotsampasakou
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria.,Computational Toxicology Group, CMS, R&D Platform Technology & Science, GSK, Park Road, Ware, Hertfordshire, SG12 0DP, UK
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria.
| |
Collapse
|
273
|
Nagarajan N, Chellam J, Kannan RR. Exploring the functional impact of mutational drift in LRRK2 gene and identification of specific inhibitors for the treatment of Parkinson disease. J Cell Biochem 2018; 119:4878-4889. [PMID: 29369408 DOI: 10.1002/jcb.26703] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 01/23/2018] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is a disorder of the central nervous system that is caused due to the death of the dopaminergic neurons in the region of the brain called substantia nigra. Mutations in LRRK2 genes are associated with disease condition and it's been reported as crucial factor for drug resistance. Identification of deleterious mutations and studying the structural and functional impact of such mutations may lead to the identification of potential selective inhibitors. In this study, we analyzed 52 PD associated mutations, among that 20 were identified as highly deleterious. The deleterious mutations G2019S and I2020T in the kinase domain were playing a key role in causing resistance to drug levedopa. Molecular docking analyses have been performed to understand the binding affinity of levodapa with LRRK2 in wild and mutant condition. Molecular docking results show that levedopa binds differentially and obtained less number of hydrogen bonds in compared with wild type LRRK2. In addition, molecular dynamics simulations were performed to study the efficacy of docked complexes and it was observed that the efficacy of the mutant complexes (G2019S-Levodopa and I2020T-Levodopa) affected in the presence of mutation. Finally, through virtual screening approach specific inhibitors SCHEMBL6473053 and SCHEMBL1278779 have been identified that could potentially inhibit LLRK2 mutants G2019S and I2020T respectively. Over all this computational investigation correlates the impact of genotypic modulation in structure and function of drug target which enhanced in the identification of precision medicine to treat PD.
Collapse
Affiliation(s)
- Nagasundaram Nagarajan
- Molecular and Nanomedicine Research Unit, Centre for Nanoscience and Nanotechnology, Sathyabama University, Chennai, Tamil Nadu, India
| | - Jaynthy Chellam
- Department of Bioinformatics, Sathyabama University, Chennai, Tamil Nadu, India
| | - Rajaretinam Rajesh Kannan
- Molecular and Nanomedicine Research Unit, Centre for Nanoscience and Nanotechnology, Sathyabama University, Chennai, Tamil Nadu, India
| |
Collapse
|
274
|
Nesbitt NM, Zheng X, Li Z, Manso JA, Yen WY, Malone LE, Ripoll-Rozada J, Pereira PJB, Mantle TJ, Wang J, Bahou WF. In silico and crystallographic studies identify key structural features of biliverdin IXβ reductase inhibitors having nanomolar potency. J Biol Chem 2018; 293:5431-5446. [PMID: 29487133 DOI: 10.1074/jbc.ra118.001803] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/23/2018] [Indexed: 12/20/2022] Open
Abstract
Heme cytotoxicity is minimized by a two-step catabolic reaction that generates biliverdin (BV) and bilirubin (BR) tetrapyrroles. The second step is regulated by two non-redundant biliverdin reductases (IXα (BLVRA) and IXβ (BLVRB)), which retain isomeric specificity and NAD(P)H-dependent redox coupling linked to BR's antioxidant function. Defective BLVRB enzymatic activity with antioxidant mishandling has been implicated in metabolic consequences of hematopoietic lineage fate and enhanced platelet counts in humans. We now outline an integrated platform of in silico and crystallographic studies for the identification of an initial class of compounds inhibiting BLVRB with potencies in the nanomolar range. We found that the most potent BLVRB inhibitors contain a tricyclic hydrocarbon core structure similar to the isoalloxazine ring of flavin mononucleotide and that both xanthene- and acridine-based compounds inhibit BLVRB's flavin and dichlorophenolindophenol (DCPIP) reductase functions. Crystallographic studies of ternary complexes with BLVRB-NADP+-xanthene-based compounds confirmed inhibitor binding adjacent to the cofactor nicotinamide and interactions with the Ser-111 side chain. This residue previously has been identified as critical for maintaining the enzymatic active site and cellular reductase functions in hematopoietic cells. Both acridine- and xanthene-based compounds caused selective and concentration-dependent loss of redox coupling in BLVRB-overexpressing promyelocytic HL-60 cells. These results provide promising chemical scaffolds for the development of enhanced BLVRB inhibitors and identify chemical probes to better dissect the role of biliverdins, alternative substrates, and BLVRB function in physiologically relevant cellular contexts.
Collapse
Affiliation(s)
| | - Xiliang Zheng
- the State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, ChangChun, Jilin 130022, China
| | | | - José A Manso
- the IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.,the i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal, and
| | | | | | - Jorge Ripoll-Rozada
- the IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.,the i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal, and
| | - Pedro José Barbosa Pereira
- the IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.,the i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal, and
| | - Timothy J Mantle
- the Department of Biochemistry, Trinity College, Dublin 2, Ireland
| | - Jin Wang
- Chemistry and Physics, State University of New York at Stony Brook, Stony Brook, New York 11794-8151,
| | | |
Collapse
|
275
|
Kumar A, Behera PC, Rangra NK, Dey S, Kant K. Computational tool for immunotoxic assessment of pyrethroids toward adaptive immune cell receptors. Pharmacogn Mag 2018; 14:124-128. [PMID: 29576712 PMCID: PMC5858232 DOI: 10.4103/pm.pm_62_17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 03/08/2017] [Indexed: 11/18/2022] Open
Abstract
Background: Pyrethroids have prominently known for their insecticidal actions worldwide, but recent reports as anticancer and antiviral applications gained a lot of interest to further understand their safety and immunotoxicity. Objective: This encouraged us to carry out our present study to evaluate the interactions of pyrethroids toward adaptive immune cell receptors. Materials and Methods: Type 1 and Type 2 pyrethroids were tested on T (CD4 and CD8) and B (CD28 and CD45) immune cell receptors using Maestro 9.3 (Schrödinger, LLC, Cambridge, USA). In addition, top-ranked tested ligands were too explored for toxicity prediction in rodents using ProTOX tool. Results: Pyrethroids (specifically type 2) such as fenvalerate (−5.534 kcal/mol: CD8), fluvalinate (−4.644 and − 4.431 kcal/mol: CD4 and CD45), and cypermethrin (−3.535 kcal/mol: CD28) have outcome in less energy or more affinity for B-cell and T-cell immune receptors which may later result in the immunosuppressive and hypersensitivity reactions. Conclusion: The current findings have uncovered that there is a further need to assess the Type 2 pyrethroids with wet laboratory experiments to understand the chemical nature of pyrethroid-induced immunotoxicity. SUMMARY Fenvalerate showed apex glide score toward CD8 immune receptor, while fluvalinate confirmed top-ranked binding with CD4 and CD45 immune proteins In addition, cypermethrin outcame in top glide score against CD28 immune receptor Top dock hits (Type 2) pyrethroids have shown probable toxicity targets toward AOFA: Amine oxidase (flavin-containing) A and PGH1: Prostaglandin G/H synthase 1, respectively.
Abbreviations used: PDB: Protein Data Bank; AOFA: Amine oxidase (flavin-containing) A; PGH 1: Prostaglandin G/H synthase 1.
Collapse
Affiliation(s)
- Anoop Kumar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Padma Charan Behera
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Naresh Kumar Rangra
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Suddhasattya Dey
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Kamal Kant
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| |
Collapse
|
276
|
Erukainure OL, Hafizur RM, Kabir N, Choudhary MI, Atolani O, Banerjee P, Preissner R, Chukwuma CI, Muhammad A, Amonsou EO, Islam MS. Suppressive Effects of Clerodendrum volubile P Beauv. [Labiatae] Methanolic Extract and Its Fractions on Type 2 Diabetes and Its Complications. Front Pharmacol 2018; 9:8. [PMID: 29449808 PMCID: PMC5799276 DOI: 10.3389/fphar.2018.00008] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 01/04/2018] [Indexed: 01/01/2023] Open
Abstract
Type 2 diabetes is the most prominent of all diabetes types, contributing to global morbidity and mortality. Availability and cost of treatment with little or no side effect especially in developing countries, remains a huge burden. This has led to the search of affordable alternative therapies especially from medicinal plants. In this study, the antidiabetic effect of the methanolic extract, dichloromethane (DCM), butanol (BuOH) and aqueous fractions of Clerodendrum volubile leaves were investigated in type 2 diabetic rats for their effect on glucose homeostasis, serum insulin level and hepatic biomarkers, lipid profile, pancreatic redox balance and Ca2+ levels, and β-cell distribution and function. The DCM was further fractionated to isolate the active compounds, biochanin and 5,7,4'-trimethoxykaempferol. They were investigated for their toxicity and ADMET properties, α-glucosidase and angiotensin I converting enzyme (ACE) inhibitory activities in silico. There were significant (p < 0.05) decrease in blood glucose, cholesterol, LDL-C, vLDL-C, triglyceride, AST and ALT levels in all treated groups, with DCM fraction showing the best activity. All treated rats showed significantly (p < 0.05) improved anti-oxidative activities. Treatment with the DCM fraction led to significant (p < 0.05) increased serum insulin and pancreatic Ca2+ levels, as well as improved β-cell distribution and function. DCM fraction also showed improved glucose tolerance. DCM fraction dose-dependently inhibited ACE activity. The toxicity class of the isolated compounds was predicted to be 5. They were also predicted to be potent inhibitors of cytochrome P (CYPs) 1A2, 2D6 and 3A4. They docked well with α-glucosidase and ACE. These results indicate the therapeutic potential of the plant against type 2 diabetes, with the DCM fraction being the most potent which may be attributed to the isolated flavones. It further suggests antihypertensive potentials of the DCM fraction. However, inhibition of CYPs by the flavones may suggest caution in usage with other prescribed drugs metabolized by these enzymes.
Collapse
Affiliation(s)
- Ochuko L. Erukainure
- Nutrition and Toxicology Division, Federal Institute of Industrial Research Oshodi, Lagos, Nigeria
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Rahman M. Hafizur
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Nurul Kabir
- Faculty of Science, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - M. Iqbal Choudhary
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Olubunmi Atolani
- Structural Bioinformatics Group, Institute for Physiology, Charité – University Medicine Berlin, Berlin, Germany
- Department of Chemistry, University of Ilorin, Ilorin, Nigeria
| | - Priyanka Banerjee
- Structural Bioinformatics Group, Institute for Physiology, Charité – University Medicine Berlin, Berlin, Germany
| | - Robert Preissner
- Structural Bioinformatics Group, Institute for Physiology, Charité – University Medicine Berlin, Berlin, Germany
| | - Chika I. Chukwuma
- Department of Food Technology, Durban University of Technology, Steve Biko Campus, Durban, South Africa
- Department of Pharmacology, University of the Free State, Bloemfontein, South Africa
| | - Aliyu Muhammad
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | - Eric O. Amonsou
- Department of Food Technology, Durban University of Technology, Steve Biko Campus, Durban, South Africa
| | - Md. Shahidul Islam
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| |
Collapse
|
277
|
Kant K, Lal UR, Ghosh M. Computational Breakthrough of Natural Lead Hits from the Genus of Arisaema against Human Respiratory Syncytial Virus. Pharmacogn Mag 2018; 13:S780-S785. [PMID: 29491633 PMCID: PMC5822500 DOI: 10.4103/pm.pm_459_16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 11/26/2016] [Indexed: 11/26/2022] Open
Abstract
Background: To date, efforts for the prevention and treatment of human respiratory syncytial virus (RSV) infection have been still vain, and there is no safe and effective clinical accepted vaccine. Arisaema genus has claimed for various traditional bioactivities, but scientific assessments are quite limited. Objective: This encouraged us to carry out our present study on around 60 phytoconstituents of different Arisaema species as a natural inhibitor against the human RSV. Materials and Methods: Selected 60 phytochemical entities were evaluated on the docking behavior of human RSV receptor (PDB: 4UCC) using Maestro 9.3 (Schrödinger, LLC, Cambridge, USA). Furthermore, kinetic properties and toxicity nature of top graded ligands were analyzed through QikProp and ProTox tools. Results: Notably, rutin (glide score: −8.49), schaftoside (glide score: −8.18) and apigenin-6,8-di-C-β-D-galactoside (glide score − 7.29) have resulted in hopeful natural lead hits with an ideal range of kinetic descriptors values. ProTox tool (oral rodent toxicity) has resulted in likely toxicity targets of apex-graded tested ligands. Conclusion: Finally, the whole efforts can be explored further as a model to confirm its anti-human RSV potential with wet laboratory experiments. SUMMARY Rutin, schaftoside, and apigenin-6,8-di-C-β-D-galactoside showed promising top hits docking profile against human respiratory syncytial virus Moreover, absorption, distribution, metabolism, excretion properties (QikProp) of top hits resulted within an ideal range of kinetic descriptors ProTox tool highlighted toxicity class ranges, LD50 values, and possible toxicity targets of apex-graded tested ligands.
Abbreviations used: RSV: Respiratory syncytial virus, PRRSV: Porcine respiratory and reproductive syndrome virus, ADME-T: Absorption, distribution, metabolism, excretion, and toxicity.
Collapse
Affiliation(s)
- Kamal Kant
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Uma Ranjan Lal
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Manik Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| |
Collapse
|
278
|
In Vitro, In Silico, and In Vivo Analyses of Novel Aromatic Amidines against Trypanosoma cruzi. Antimicrob Agents Chemother 2018; 62:AAC.02205-17. [PMID: 29203486 DOI: 10.1128/aac.02205-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 11/20/2017] [Indexed: 12/16/2022] Open
Abstract
Five bis-arylimidamides were assayed as anti-Trypanosoma cruzi agents by in vitro, in silico, and in vivo approaches. None were considered to be pan-assay interference compounds. They had a favorable pharmacokinetic landscape and were active against trypomastigotes and intracellular forms, and in combination with benznidazole, they gave no interaction. The most selective agent (28SMB032) tested in vivo led to a 40% reduction in parasitemia (0.1 mg/kg of body weight/5 days intraperitoneally) but without mortality protection. In silico target fishing suggested DNA as the main target, but ultrastructural data did not match.
Collapse
|
279
|
Zheng M, Zhao J, Cui C, Fu Z, Li X, Liu X, Ding X, Tan X, Li F, Luo X, Chen K, Jiang H. Computational chemical biology and drug design: Facilitating protein structure, function, and modulation studies. Med Res Rev 2018; 38:914-950. [DOI: 10.1002/med.21483] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/13/2017] [Accepted: 12/15/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Mingyue Zheng
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Jihui Zhao
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Chen Cui
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Zunyun Fu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Xutong Li
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Xiaohong Liu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
- School of Life Science and Technology; ShanghaiTech University; Shanghai China
| | - Xiaoyu Ding
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Xiaoqin Tan
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Fei Li
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
- Department of Chemistry, College of Sciences; Shanghai University; Shanghai China
| | - Xiaomin Luo
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| | - Kaixian Chen
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
- School of Life Science and Technology; ShanghaiTech University; Shanghai China
| | - Hualiang Jiang
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai China
| |
Collapse
|
280
|
Pires DEV, Kaminskas LM, Ascher DB. Prediction and Optimization of Pharmacokinetic and Toxicity Properties of the Ligand. Methods Mol Biol 2018; 1762:271-284. [PMID: 29594777 DOI: 10.1007/978-1-4939-7756-7_14] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
A crucial factor for the approval and success of any drug is how it behaves in the body. Many drugs, however, do not reach the market due to poor efficacy or unacceptable side effects. It is therefore important to take these into consideration early in the drug development process, both in the prioritization of potential hits, and optimization of lead compounds. In silico approaches offer a cost and time-effective approach to rapidly screen and optimize pharmacokinetic and toxicity properties. Here we demonstrate the use of the comprehensive analysis system pkCSM, to allow early identification of potential problems, prioritization of hits, and optimization of leads.
Collapse
Affiliation(s)
| | - Lisa M Kaminskas
- School of Biomedical Sciences, University of Queensland, St. Lucia, QLD, Australia
| | - David B Ascher
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia.
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
| |
Collapse
|
281
|
Abstract
The concept of chemical similarity has many applications in several fields of cheminformatics. One common use of chemical similarity measurements, based on the principle that similar molecules have similar properties, is in the context of the read-across approach, where estimates of a specific endpoint for a chemical are obtained starting from experimental data available from highly similar compounds.This chapter reports an implementation of chemical similarity and the analysis of multiple combinations of binary fingerprints and similarity metrics in the context of the read-across technique.This analysis demonstrates that the classical similarity measurements can be improved with a generalizable model of similarity. The approach presented here has been implemented in two open-source software tools for computational toxicology (CAESAR and VEGA).
Collapse
Affiliation(s)
- Matteo Floris
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.
- IRGB - CNR, National Research Council, Institute of Genetics and Biomedical Research, Monserrato, CA, Italy.
| | - Stefania Olla
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
282
|
Nasab RR, Mansourian M, Hassanzadeh F, Shahlaei M. Exploring the interaction between epidermal growth factor receptor tyrosine kinase and some of the synthesized inhibitors using combination of in-silico and in-vitro cytotoxicity methods. Res Pharm Sci 2018; 13:509-522. [PMID: 30607149 PMCID: PMC6288988 DOI: 10.4103/1735-5362.245963] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Quinazoline derivatives are potent inhibitors of human epidermal growth factor receptor (EGFR) as anticancer agents. In this study, the cytotoxic effects of a new series of synthesized quinazoline derivatives were evaluated using MTT assay against MCF-7 and HT-29 cell lines. Using molecular docking, the binding modes of all compounds were analyzed at the binding site of EGFR. Based on the results, the compounds L1, L2, L4, L5, L6, L7, L10, L15, and L18 may be promising EGFR inhibitors based on docking score and hydrogen bonds. Consistent with the experimental data, Met769 is recognized as a key residue in the binding of potential inhibitors. According to the MTT cytotoxicity assays, Lipinski's rule of five (RO5), absorption, distribution, metabolism, excretion, and toxicity (ADMET) parameters, and docking studies, three compounds L4, L15, and L10 with IC50 values of 80, 60, and 1 μM against the MCF-7 were selected for further comparative assessments. The dynamics of free EGFR, and selected ligand-EGFR complexes were investigated using molecular dynamics (MD) simulation studies. The results indicated that the three compounds bound to EGFR active site in a stable manner during the simulation through the formation of new hydrogen bonds with Phe699, Leu694, Gly700, Lys721, Met769, Arg817, and Asp831 with the superiority of compound L15. These features can promote future drug candidate designing to produce better derivatives in the search for the anticancer agents.
Collapse
Affiliation(s)
- Rezvan Rezaee Nasab
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Lorestan University of Medical Sciences, Khorramabad, I.R. Iran.,Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mahboubeh Mansourian
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, I.R. Iran.,Department of Pharmacology, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, I.R. Iran
| | - Farshid Hassanzadeh
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mohsen Shahlaei
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, I.R. Iran
| |
Collapse
|
283
|
Sarabia-Sánchez MJ, Trejo-Soto PJ, Velázquez-López JM, Carvente-García C, Castillo R, Hernández-Campos A, Avitia-Domínguez C, Enríquez-Mendiola D, Sierra-Campos E, Valdez-Solana M, Salas-Pacheco JM, Téllez-Valencia A. Novel Mixed-Type Inhibitors of Protein Tyrosine Phosphatase 1B. Kinetic and Computational Studies. Molecules 2017; 22:molecules22122262. [PMID: 29261102 PMCID: PMC6150025 DOI: 10.3390/molecules22122262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/13/2017] [Accepted: 12/16/2017] [Indexed: 11/21/2022] Open
Abstract
The Atlas of Diabetes reports 415 million diabetics in the world, a number that has surpassed in half the expected time the twenty year projection. Type 2 diabetes is the most frequent form of the disease; it is characterized by a defect in the secretion of insulin and a resistance in its target organs. In the search for new antidiabetic drugs, one of the principal strategies consists in promoting the action of insulin. In this sense, attention has been centered in the protein tyrosine phosphatase 1B (PTP1B), a protein whose overexpression or increase of its activity has been related in many studies with insulin resistance. In the present work, a chemical library of 250 compounds was evaluated to determine their inhibition capability on the protein PTP1B. Ten molecules inhibited over the 50% of the activity of the PTP1B, the three most potent molecules were selected for its characterization, reporting Ki values of 5.2, 4.2 and 41.3 µM, for compounds 1, 2, and 3, respectively. Docking and molecular dynamics studies revealed that the three inhibitors made interactions with residues at the secondary binding site to phosphate, exclusive for PTP1B. The data reported here support these compounds as hits for the design more potent and selective inhibitors against PTP1B in the search of new antidiabetic treatment.
Collapse
Affiliation(s)
- Marie Jazmín Sarabia-Sánchez
- Facultad de Medicina y Nutrición, Universidad Juárez del Estado de Durango, Av. Universidad y Fanny Anitúa S/N, Durango, Durango C.P. 34000, Mexico.
| | - Pedro Josué Trejo-Soto
- Facultad de Química, Departamento de Farmacia, Universidad Nacional Autónoma de México, Ciudad de México C.P. 04510, Mexico.
| | - José Miguel Velázquez-López
- Facultad de Química, Departamento de Farmacia, Universidad Nacional Autónoma de México, Ciudad de México C.P. 04510, Mexico.
| | - Carlos Carvente-García
- Facultad de Química, Departamento de Farmacia, Universidad Nacional Autónoma de México, Ciudad de México C.P. 04510, Mexico.
| | - Rafael Castillo
- Facultad de Química, Departamento de Farmacia, Universidad Nacional Autónoma de México, Ciudad de México C.P. 04510, Mexico.
| | - Alicia Hernández-Campos
- Facultad de Química, Departamento de Farmacia, Universidad Nacional Autónoma de México, Ciudad de México C.P. 04510, Mexico.
| | - Claudia Avitia-Domínguez
- Facultad de Medicina y Nutrición, Universidad Juárez del Estado de Durango, Av. Universidad y Fanny Anitúa S/N, Durango, Durango C.P. 34000, Mexico.
| | - Daniel Enríquez-Mendiola
- Facultad de Medicina y Nutrición, Universidad Juárez del Estado de Durango, Av. Universidad y Fanny Anitúa S/N, Durango, Durango C.P. 34000, Mexico.
| | - Erick Sierra-Campos
- Facultad de Ciencias Químicas, Universidad Juárez del Estado de Durango, Av. Artículo 123 S/N Fracc. Filadelfia, Gómez Palacio, Durango C.P. 35010, Mexico.
| | - Mónica Valdez-Solana
- Facultad de Ciencias Químicas, Universidad Juárez del Estado de Durango, Av. Artículo 123 S/N Fracc. Filadelfia, Gómez Palacio, Durango C.P. 35010, Mexico.
| | - José Manuel Salas-Pacheco
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Av. Universidad S/N, Durango, Durango C.P. 34000, Mexico.
| | - Alfredo Téllez-Valencia
- Facultad de Medicina y Nutrición, Universidad Juárez del Estado de Durango, Av. Universidad y Fanny Anitúa S/N, Durango, Durango C.P. 34000, Mexico.
| |
Collapse
|
284
|
Ellul C, Shoemake C. Design of Novel Compounds with the Potential of Dual PPARγ/α Modulation for the Management of Metabolic Syndrome. NUCLEAR RECEPTOR RESEARCH 2017. [DOI: 10.11131/2017/101311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
285
|
Dacryodes edulis enhances antioxidant activities, suppresses DNA fragmentation in oxidative pancreatic and hepatic injuries; and inhibits carbohydrate digestive enzymes linked to type 2 diabetes. Biomed Pharmacother 2017; 96:37-47. [DOI: 10.1016/j.biopha.2017.09.106] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 12/31/2022] Open
|
286
|
Caffeine – rich infusion from Cola nitida (kola nut) inhibits major carbohydrate catabolic enzymes; abates redox imbalance; and modulates oxidative dysregulated metabolic pathways and metabolites in Fe2+-induced hepatic toxicity. Biomed Pharmacother 2017; 96:1065-1074. [DOI: 10.1016/j.biopha.2017.11.120] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/05/2017] [Accepted: 11/27/2017] [Indexed: 12/11/2022] Open
|
287
|
Zhou X, Li H, Shi Z, Gao S, Wei S, Li K, Wang J, Li J, Wang R, Gong M, Zhao Y, Xiao X. Inhibition activity of a traditional Chinese herbal formula Huang-Lian-Jie-Du-Tang and its major components found in its plasma profile on neuraminidase-1. Sci Rep 2017; 7:15549. [PMID: 29138445 PMCID: PMC5686190 DOI: 10.1038/s41598-017-15733-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 10/31/2017] [Indexed: 12/25/2022] Open
Abstract
Huang-Lian-Jie-Du-Tang (HLJDT), a traditional formula with four TCM herbs, has been used for hundred years for different diseases. The current study aimed to assess the inhibitory activity of HLJDT against H1N1 neuraminidase (NA-1), and identify potent NA-1 inhibitors from its plasma profile. The in vitro NA-1 study has shown that the water extract of HLJDT potently inhibited NA-1 (IC50 = 112.6 μg/ml; Ki = 55.6 μg/ml) in a competitive mode. The IC50 values of the water extracts of its four herbs were as follows: Coptidis Rhizoma, 96.1 μg/ml; Phellodendri Chinensis Cortex, 108.6 μg/ml; Scutellariae Radix, 303.5 μg/ml; Gardeniae Fructus, 285.0 μg/ml. Thirteen compounds found in the plasma profile of HLJDT were also identified as potent NA-1 inhibitors, which included jatrorrhizine, palmatine, epiberberine, geniposide, oroxylin A, berberine, coptisine, baicalein, wogonoside, phellodendrine, wogonin, oroxylin A-7-O-glucuronide and baicalin (sorted in ascending order by their IC50 values). Their inhibitory activities were consistent with molecular docking analysis when considering crystallographic water molecules in the ligand-binding pocket of NA-1. Our current findings suggested that HLJDT can be used as a complementary medicine for H1N1 infection and its potent active compounds can be developed as NA-1 inhibitors.
Collapse
Affiliation(s)
- Xuelin Zhou
- Department of Pharmacy, 302 Military Hospital of China, Beijing, People's Republic of China
| | - Haotian Li
- Department of Pharmacy, 302 Military Hospital of China, Beijing, People's Republic of China
| | - Zhilong Shi
- China Military Institute of Chinese Medicine, 302 Military Hospital of China, Beijing, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Sijia Gao
- Department of Pharmacy, 302 Military Hospital of China, Beijing, People's Republic of China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Shizhang Wei
- Department of Pharmacy, 302 Military Hospital of China, Beijing, People's Republic of China
| | - Kun Li
- Department of Pharmacy, 302 Military Hospital of China, Beijing, People's Republic of China
| | - Jiabo Wang
- China Military Institute of Chinese Medicine, 302 Military Hospital of China, Beijing, People's Republic of China
- Integrative Medical Center, 302 Military Hospital of China, Beijing, People's Republic of China
| | - Jianyu Li
- Integrative Medical Center, 302 Military Hospital of China, Beijing, People's Republic of China
| | - Ruilin Wang
- Integrative Medical Center, 302 Military Hospital of China, Beijing, People's Republic of China
| | - Man Gong
- Integrative Medical Center, 302 Military Hospital of China, Beijing, People's Republic of China
| | - Yanling Zhao
- Department of Pharmacy, 302 Military Hospital of China, Beijing, People's Republic of China.
| | - Xiaohe Xiao
- China Military Institute of Chinese Medicine, 302 Military Hospital of China, Beijing, People's Republic of China
- Integrative Medical Center, 302 Military Hospital of China, Beijing, People's Republic of China
| |
Collapse
|
288
|
Banerjee A, Pal A, Pal D, Mitra P. Ebolavirus interferon antagonists—protein interaction perspectives to combat pathogenesis. Brief Funct Genomics 2017; 17:392-401. [DOI: 10.1093/bfgp/elx034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
289
|
Schuler J, Hudson ML, Schwartz D, Samudrala R. A Systematic Review of Computational Drug Discovery, Development, and Repurposing for Ebola Virus Disease Treatment. Molecules 2017; 22:E1777. [PMID: 29053626 PMCID: PMC6151658 DOI: 10.3390/molecules22101777] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/16/2017] [Accepted: 09/19/2017] [Indexed: 12/30/2022] Open
Abstract
Ebola virus disease (EVD) is a deadly global public health threat, with no currently approved treatments. Traditional drug discovery and development is too expensive and inefficient to react quickly to the threat. We review published research studies that utilize computational approaches to find or develop drugs that target the Ebola virus and synthesize its results. A variety of hypothesized and/or novel treatments are reported to have potential anti-Ebola activity. Approaches that utilize multi-targeting/polypharmacology have the most promise in treating EVD.
Collapse
Affiliation(s)
- James Schuler
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, USA.
| | - Matthew L Hudson
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, USA.
| | - Diane Schwartz
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, USA.
| | - Ram Samudrala
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, USA.
| |
Collapse
|
290
|
Grecco SS, Costa-Silva TA, Jerz G, de Sousa FS, Londero VS, Galuppo MK, Lima ML, Neves BJ, Andrade CH, Tempone AG, Lago JHG. Neolignans from leaves of Nectandra leucantha (Lauraceae) display in vitro antitrypanosomal activity via plasma membrane and mitochondrial damages. Chem Biol Interact 2017; 277:55-61. [PMID: 28864277 DOI: 10.1016/j.cbi.2017.08.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/13/2017] [Accepted: 08/28/2017] [Indexed: 12/28/2022]
Abstract
Chagas disease is a neglected tropical disease, caused by the protozoan parasite Trypanosoma cruzi, which affects more than eight million people in Tropical and Subtropical countries especially in Latin America. Current treatment is limited to nifurtimox and benznidazole, both with reduced effectiveness and high toxicity. In this work, the n-hexane extract from leaves of Nectandra leucantha (Lauraceae) displayed in vitro antitrypanosomal activity against T. cruzi. Using several chromatographic steps, four related neolignans were isolated and chemically characterized as dehydrodieugenol B (1), 1-(8-propenyl)-3-[3'-methoxy-1'-(8-propenyl)-phenoxy]-4,5-dimethoxybenzene (2), 1-[(7S)-hydroxy-8-propenyl]-3-[3'-methoxy-1'-(8'-propenyl)-phenoxy]-4-hydroxy-5-methoxybenzene (3), and 1-[(7S)-hydroxy-8-propenyl]-3-[3'-methoxy-1'-(8'-propenyl)-phenoxy]-4,5-dimethoxybenzene (4). These compounds were tested against intracellular amastigotes and extracellular trypomastigotes of T. cruzi and for mammalian cytotoxicity. Neolignan 4 showed the higher selectivity index (SI) against trypomastigotes (>5) and amastigotes (>13) of T. cruzi. The investigation of the mechanism of action demonstrated that neolignan 4 caused substantial alteration of the plasma membrane permeability, together with mitochondrial dysfunctions in trypomastigote forms. In silico studies of pharmacokinetics and toxicity (ADMET) properties predicted that all compounds were non-mutagenic, non-carcinogenic, non-genotoxic, weak hERG blockers, with acceptable volume of distribution (1.66-3.32 L/kg), and low rodent oral toxicity (LD50 810-2200 mg/kg). Considering some clinical events of cerebral Chagas disease, the compounds also demonstrated favorable properties, such as blood-brain barrier penetration. Unfavorable properties were also predicted as high promiscuity for P450 isoforms, high plasma protein binding affinity (>91%), and moderate-to-low oral bioavailability. Finally, none of the isolated neolignans was predicted as interference compounds (PAINS). Considering the promising chemical and biological properties of the isolated neolignans, these compounds could be used as starting points to develop new lead compounds for Chagas disease.
Collapse
Affiliation(s)
- Simone S Grecco
- Center of Natural Sciences and Humanities, Federal University of ABC, Santo Andre, SP, 09210-180, Brazil; Institute of Food Chemistry, Technische Universität Braunschweig, Braunschweig, 38106, Germany; Biotechnology and Innovation in Health Program, Anhanguera University of São Paulo, São Paulo, SP, 05145-200, Brazil
| | - Thais A Costa-Silva
- Center of Natural Sciences and Humanities, Federal University of ABC, Santo Andre, SP, 09210-180, Brazil
| | - Gerold Jerz
- Institute of Food Chemistry, Technische Universität Braunschweig, Braunschweig, 38106, Germany
| | - Fernanda S de Sousa
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, SP, 09972-270, Brazil
| | - Vinicius S Londero
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, SP, 09972-270, Brazil
| | - Mariana K Galuppo
- Centre for Parasitology and Mycology, Instituto Adolfo Lutz, São Paulo, SP, 01246-902, Brazil
| | - Marta L Lima
- Centre for Parasitology and Mycology, Instituto Adolfo Lutz, São Paulo, SP, 01246-902, Brazil; Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, SP, 05403-000, Brazil
| | - Bruno J Neves
- LabMol, Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiânia, GO, 74605-170, Brazil; Postgraduate Program in Society, Technology and Environment, Unievangelica University Center, Anápolis, GO, 75083-515, Brazil
| | - Carolina H Andrade
- LabMol, Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiânia, GO, 74605-170, Brazil
| | - Andre G Tempone
- Centre for Parasitology and Mycology, Instituto Adolfo Lutz, São Paulo, SP, 01246-902, Brazil
| | - João Henrique G Lago
- Center of Natural Sciences and Humanities, Federal University of ABC, Santo Andre, SP, 09210-180, Brazil.
| |
Collapse
|
291
|
Singh P, Talwar P. Exploring putative inhibitors of Death Associated Protein Kinase 1 (DAPK1) via targeting Gly-Glu-Leu (GEL) and Pro-Glu-Asn (PEN) substrate recognition motifs. J Mol Graph Model 2017; 77:153-167. [PMID: 28858643 DOI: 10.1016/j.jmgm.2017.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 08/01/2017] [Accepted: 08/02/2017] [Indexed: 02/03/2023]
Abstract
Recently, a new signaling complex Death Associated Protein Kinase 1 (DAPK1) ̶ N-methyl-D-aspartate receptor subtype 2B (NMDAR2B or NR2B) engaged in the neuronal death cascade was identified and it was found that after stroke injury, N-methyl-D-aspartate glutamate (NMDA) receptors interact with DAPK1 through NR2B subunit and lead to excitotoxicity via over-activation of NMDA receptors. An acute brain injury, such as stroke, is a serious life-threatening medical condition which occurs due to poor blood supply to the brain and further leads to neuronal cell death. During a stroke, activated DAPK1 migrates towards the extra-synaptic site and binds to NR2B subunit of NMDA receptor. It is this DAPK1-NR2B interaction that arbitrates the pathological processes like apoptosis, necrosis, and autophagy of neuronal cells observed in stroke injury, hence we aimed to inhibit this vital interaction to prevent neuronal damage. In the present study, using PubChem database, we applied an integrative approach of virtual screening and molecular dynamic simulations and identified a potential lead compound 11 that interrupts DAPK1-NR2B interaction by competing with both ATP and substrate for their binding sites on DAPK1. This inhibitor was found potent and considerably selective to DAPK1 as it made direct contact with the ATP binding sites as well as substrate recognition motifs: Gly-Glu-Leu (GEL) and Pro-Glu-Asn (PEN). Further in vitro and in vivo experiments are demanded to validate the efficacy of compound 11 nevertheless, it can be considered as suitable starting point for designing DAPK1 inhibitors.
Collapse
Affiliation(s)
- Pratibha Singh
- Apoptosis and Cell Survival Research Laboratory, Department of Bio-Sciences, School of Bio Sciences and Technology, VIT University, Vellore, Tamil Nadu 632014, India
| | - Priti Talwar
- Apoptosis and Cell Survival Research Laboratory, Department of Bio-Sciences, School of Bio Sciences and Technology, VIT University, Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
292
|
Murahari M, Prakash KV, Peters GJ, Mayur YC. Acridone-pyrimidine hybrids- design, synthesis, cytotoxicity studies in resistant and sensitive cancer cells and molecular docking studies. Eur J Med Chem 2017; 139:961-981. [PMID: 28886509 DOI: 10.1016/j.ejmech.2017.08.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/26/2017] [Accepted: 08/08/2017] [Indexed: 12/15/2022]
Abstract
Hybrid systems of acridones with substituted pyrimidines were designed with an objective of discovering next generation anticancer agents targeting multiple mechanisms in the cancer cell. Hybrid compounds were synthesized by simple and convenient methods in the lab, characterized by NMR and Mass spectral methods and screened for cytotoxicity against A549 (lung), Hela (cervical), MCF7 (breast) and MDA-MB-231 (breast) cancer cell lines respectively. Evaluation of compounds for cell proliferation identified active compounds 11b, 11d and 11h against MCF7, MDA-MB-231 and A549 cell lines. Further absorption titrations with CT-DNA and gel electrophoresis identified that hybrid molecules displayed anticancer activity partly by DNA intercalation. Also further results of western blotting assay with Akt kinase identified that hybrid compounds have the ability to inhibit the Akt kinase activity and induce apoptosis, with ABCC1 suggests that active compounds too have the ability to modulate multidrug resistance (MDR) associated with ABCC1/MRP1. Selective Akt1 kinase assay have identified 11a, 11b, 11d and 11h as potential inhibitors. Molecular docking studies identified the orientation and binding interactions at the active site of Akt1 and DNA. Compounds 12e and 12f have shown good cytotoxicity profile against lung cancer cell lines of sensitive and resistant type. Acute toxicity study of compound 12f at the dose of 5000 mg/kg has identified no signs of clinical toxicity. Prediction of ADMET properties and oral toxicity of the drug likeness features of new hybrid systems were carried out using software's. This experimental data suggests that hybrid systems of acridone with substituted pyrimidines can be taken as a lead for the design of efficient inhibitors and active compounds which can be taken up for further studies.
Collapse
Affiliation(s)
- Manikanta Murahari
- Department of Pharmaceutical Chemistry, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400 056, India
| | | | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Y C Mayur
- Department of Pharmaceutical Chemistry, SPP School of Pharmacy & Technology Management, SVKM's NMIMS- Deemed to be University, Mumbai 400 056, India.
| |
Collapse
|
293
|
Erukainure OL, Mesaik MA, Atolani O, Muhammad A, Chukwuma CI, Islam MS. Pectolinarigenin from the leaves of Clerodendrum volubile shows potent immunomodulatory activity by inhibiting T - cell proliferation and modulating respiratory oxidative burst in phagocytes. Biomed Pharmacother 2017; 93:529-535. [PMID: 28686966 DOI: 10.1016/j.biopha.2017.06.060] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 01/07/2023] Open
Abstract
There have been increasing interest in the use of plant-derived substance as immunomodulators for the treatment and management of inflammatory ailments. Clerodendrum volubile, a leafy vegetable is known for its folkloric applications in the treatments of several inflammatory related ailments, but with little scientific evidence. This study reports the isolation, structure elucidation and in vitro immunomodulatory potentials of pectolinarigenin from C. volubile leaves. The immunomodulatory potentials of the crude methanolic extract and fractions [n-hexane (Hex), dichloromethane (DCM), ethyl acetate (EtOAc) and n - butanol (BuOH)] were investigated on whole blood, neutrophil and macrophage phagocytic respiratory burst using luminol-amplified chemiluminescence technique. DCM fraction showed higher inhibitory activity on respiratory burst, indicating high suppressive immunomodulatory potency. The DCM fraction was further fractionated using a gravity column chromatography loaded with silica gel. The column was eluted with mixtures of Hex and DCM (92.5:7.5) in increasing order of polarity up to Hex: DCM (88:12) to afford 5,7-Dihydroxy-6,4'-dimethoxyflavone (pectolinarigenin). The structure of the compound was established using data obtained from 1H- and 13C NMR spectroscopies and mass spectrometry. The isolated flavone was investigated for its inhibitory activity of neutrophil phagocytes respiratory burst as well as T - Cell proliferation. The compound exhibited significant activities (at p <0.05) indicating high suppressive immunomodulatory potency. The potent suppressive effect of pectolinarigenin on polymorphonuclear neutrophils (PMNs) respiratory oxidative burst and T - cell proliferation suggests an immunomodulatory potential and pathway of the flavonoid.
Collapse
Affiliation(s)
- Ochuko L Erukainure
- Nutrition Toxicology Division, Federal Institute of Industrial Research, Lagos, Nigeria; Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan; Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, (Westville Campus), Durban 4000, South Africa.
| | - M Ahmed Mesaik
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan; Faculty of Medicine, University of Tabuk, P.O. Box 741, Tabuk 71491 Saudi Arabia
| | | | - Aliyu Muhammad
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan; Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | - Chika I Chukwuma
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, (Westville Campus), Durban 4000, South Africa
| | - Md Shahidul Islam
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, (Westville Campus), Durban 4000, South Africa
| |
Collapse
|
294
|
Mohan S, Hemachandran H, Sneha P, George Priya Doss C, Godwin Christopher J, Jayaraman G, Ramamoorthy S. Structural insights into the binding mode and conformational changes of BSA induced by bixin and crocin. J Biomol Struct Dyn 2017; 36:2085-2098. [DOI: 10.1080/07391102.2017.1342565] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Sankari Mohan
- School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - Hridya Hemachandran
- School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - P. Sneha
- School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - C. George Priya Doss
- School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - J. Godwin Christopher
- School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - Gurunathan Jayaraman
- School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - Siva Ramamoorthy
- School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| |
Collapse
|
295
|
Ghafari S, Komeilian M, Hashemi MS, Oushani S, Rigi G, Rashidieh B, Yarahmadi K, Khoddam F. Molecular docking based screening of Listeriolysin-O for improved inhibitors. Bioinformation 2017; 13:160-163. [PMID: 28690383 PMCID: PMC5498783 DOI: 10.6026/97320630013160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/23/2017] [Accepted: 04/23/2017] [Indexed: 12/30/2022] Open
Abstract
Listeriolysine-O (LLO) is a 50KDa protein responsible for Listeria monocytogenes pathogenicity. The structure of LLO (PDB ID: 4CDB) with domains D1 to D4 is known. Therefore, it is of interest to identify conserved regions among LLO variants for destabilizing oligomerization (50 mer complex) of its monomers using appropriate inhibitors. Therefore, it is of interest to identify suitable inhibitors for inhibiting LLO. Previous reports suggest the use of flavanoids like compounds for inhibiting LLO. Our interest is to identify improved compounds to destabilize LLO oligomerization. We used a library (Zinc database) containing 200,000 drug-like compounds against LLO using molecular docking based screening. This resulted in five hits that were further analyzed for pharmacological properties. The hit #1 (2-methyloctadecane- 1, 3, 4-triol) was further refined using appropriate modifications for creating a suitable pharmacophore model LLO inhibition. The modified compound (1-(4-Cyclopent-3-enyl-6, 7-dihydroxy-8-hydroxymethyl-nona-2, 8-dienylideneamino)-penta-1,4-dien-3-one) shows fitting binding properties with LLO with no undesirable pharmacological properties such as toxicity.
Collapse
Affiliation(s)
| | | | | | | | - Garshasb Rigi
- Department of Biology, Faculty of Science, Behbahan Khatam Alanbia University of Technology, Behbahan, Iran
| | | | | | | |
Collapse
|
296
|
Zolfaghari N. Competitive rational inhibitor design to 4-maleylaceto-acetate isomerase. Bioinformation 2017; 13:140-143. [PMID: 28690379 PMCID: PMC5498779 DOI: 10.6026/97320630013140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 05/01/2017] [Indexed: 11/23/2022] Open
Abstract
Tyrosinemia type I is the result of genetic disorder in fomaryl acetoacetase gene that leads to 4-fumaryl acetoacetate accumulation. The current treatment for tyrosinemia type I is nitisinone that inhibits 4-hydroxyphenyl pyruvic dioxygenase in competitive manner. In the present study, we have designed two theoretical chemicals, which could inhibit the direct enzyme responsible for fumarylacetoacetate formation. Subset 2_p.0.5 from Zinc database was screened by PyRx software using a Lamarckian genetic algorithm as the scoring function for docking. Top nine successive hits were selected for further pharmacological analysis and finally the new designed ligands RD6-2 (3Z)- 1,3-Butadiene-1,1,2,4-tetrol and RD-7-1 ((Z)-3-[4-Hydroxy-1-(hydroxymethyl)cyclohexyl]-2-propene-1,2-diol could pass PhysChem, FAFDrugs and AdmetSAR filter. The designed ligands were non-substrate and non-inhibitor of CYP450 and nontoxic in AMES test. LD50 of RD-6-2 was 793mg/kg with the toxicity class of four and The LD50 of RD-7-1 was calculated as 5000mg/kg within the toxicity class of five. The designed molecules are introduced as the new theoretical small molecules, which can theoretically inhibit 4- maleylacetoacetate isomerase in a competitive manner.
Collapse
Affiliation(s)
- Narges Zolfaghari
- National institute of genetic engineering and biotechnology, Tehran, Iran
| |
Collapse
|
297
|
Schrey AK, Nickel-Seeber J, Drwal MN, Zwicker P, Schultze N, Haertel B, Preissner R. Computational prediction of immune cell cytotoxicity. Food Chem Toxicol 2017; 107:150-166. [PMID: 28558974 DOI: 10.1016/j.fct.2017.05.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 05/11/2017] [Accepted: 05/19/2017] [Indexed: 12/11/2022]
Abstract
Immunotoxicity, defined as adverse effects of xenobiotics on the immune system, is gaining increasing attention in the approval process of industrial chemicals and drugs. In-vivo and ex-vivo experiments have been the gold standard in immunotoxicity assessment so far, so the development of in-vitro and in-silico alternatives is an important issue. In this paper we describe a widely applicable, easy-to use computational approach which can serve as an initial immunotoxicity screen of new chemical entities. Molecular fingerprints describing chemical structure were used as parameters in a machine-learning approach based on the Naïve-Bayes learning algorithm. The model was trained using blood-cell growth inhibition data from the NCI database and validated externally with several in-house and literature-derived data sets tested in cytotoxicity assays on different types on immune cells. Both cross-validations and external validations resulted in areas under the receiver operator curves (ROC/AUC) of 75% or higher. The classification of the validation data sets occurred with excellent specificities and fair to excellent selectivities, depending on the data set. This means that the probability of actual immunotoxicity is very high for compounds classified as immunotoxic, while the fraction of false negative predictions might vary. Thus, in a multistep immunotoxicity screening scheme, the classification as immunotoxic can be accepted without additional confirmation, while compounds classified as not immunotoxic will have to be subjected to further investigation.
Collapse
Affiliation(s)
- Anna K Schrey
- Charité - University Medicine Berlin, Institute for Physiology and ECRC, Berlin, Germany
| | - Janette Nickel-Seeber
- Charité - University Medicine Berlin, Institute for Physiology and ECRC, Berlin, Germany
| | - Malgorzata N Drwal
- Charité - University Medicine Berlin, Institute for Physiology and ECRC, Berlin, Germany
| | - Paula Zwicker
- University of Greifswald, Institute of Pharmacy, Greifswald, Germany
| | - Nadin Schultze
- University of Greifswald, Institute of Pharmacy, Greifswald, Germany
| | - Beate Haertel
- University of Greifswald, Institute of Pharmacy, Greifswald, Germany
| | - Robert Preissner
- Charité - University Medicine Berlin, Institute for Physiology and ECRC, Berlin, Germany.
| |
Collapse
|
298
|
Thirumal Kumar D, Lavanya P, George Priya Doss C, Tayubi IA, Naveen Kumar DR, Francis Yesurajan I, Siva R, Balaji V. A Molecular Docking and Dynamics Approach to Screen Potent Inhibitors Against Fosfomycin Resistant Enzyme in Clinical Klebsiella pneumoniae. J Cell Biochem 2017; 118:4088-4094. [PMID: 28409871 DOI: 10.1002/jcb.26064] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/13/2017] [Indexed: 01/13/2023]
Abstract
Klebsiella pneumoniae, BA6753 was cultured from a patient in the Clinical Microbiology Laboratory of Christian Medical College. K. pneumoniae, BA6753 has a multidrug resistance plasmid encoding novel FosA variant-7, fosfomycin resistance enzyme. Minimal side effects and a wide range of bactericidal activity of fosfomycin have resulted in its expanded clinical use that prompts the rise of fosfomycin-resistant strains. At present, there are no effective inhibitors available to conflict the FosA-medicated fosfomycin resistance. To develop effective FosA inhibitors, it is crucial to understand the structural and dynamic properties of resistance enzymes. Hence, the present study focuses on the identification of potent inhibitors that can effectively bind to the fosfomycin resistance enzyme, thus predispose the target to inactivate by the second antibiotic. Initially, a series of active compounds were screened against the resistant enzyme, and the binding affinities were confirmed using docking simulation analysis. For efficient activity, the binding affinity of the resistance enzyme ought to be high with the inhibitor than the fosfomycin drug. Consequently, the enzyme-ligand complex which showed higher binding affinity than the fosfomycin was employed for subsequent analysis. The stability of the top scoring enzyme-ligand complex was further validated using molecular dynamics simulation studies. On the whole, we presume that the compound 19583672 demonstrates a higher binding affinity for the resistance enzyme comparing to other compounds and fosfomycin. We believe that further enhancement of the lead compound can serve as a potential inhibitor against resistance enzyme in drug discovery process. J. Cell. Biochem. 118: 4088-4094, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- D Thirumal Kumar
- School of Biosciences and Technology, VIT University, Vellore, 632014, India
| | - P Lavanya
- Department of Clinical Microbiology, Christian Medical College, Vellore, 632004, India
| | - C George Priya Doss
- School of Biosciences and Technology, VIT University, Vellore, 632014, India
| | - Iftikhar Aslam Tayubi
- Faculty of Computing and Information Technology, King Abdulaziz University, Rabigh, 21911, Saudi Arabia
| | - D R Naveen Kumar
- Department of Clinical Microbiology, Christian Medical College, Vellore, 632004, India
| | - I Francis Yesurajan
- Department of Clinical Microbiology, Christian Medical College, Vellore, 632004, India
| | - R Siva
- School of Biosciences and Technology, VIT University, Vellore, 632014, India
| | - V Balaji
- Department of Clinical Microbiology, Christian Medical College, Vellore, 632004, India
| |
Collapse
|
299
|
Nagasundaram N, Wilson Alphonse CR, Samuel Gnana PV, Rajaretinam RK. Molecular Dynamics Validation of Crizotinib Resistance to ALK Mutations (L1196M and G1269A) and Identification of Specific Inhibitors. J Cell Biochem 2017; 118:3462-3471. [PMID: 28332225 DOI: 10.1002/jcb.26004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/20/2017] [Indexed: 11/05/2022]
Abstract
Anaplastic lymphoma kinase (ALK) positive non-small cell lung cancer (NSCLC) patients are mostly treated with ALK tyrosine kinase inhibitors (TKIs). Crizotinib is the first generation ALK inhibitor practiced as a primary chemo to combat cancer cells followed by second generation inhibitor ceritinib which are effective against crizotinib resistant ALK mutations. However, patients treated with these drugs invariably relapsed because of the development of new drug resistance mutations. In this study we explored the crizotinib resistance in the presence of ALK mutations L1196M and G1269A through molecular dynamics simulation studies. Further mutation specific inhibitors CID 71748211 and CID 71728095 were identified to potentially inhibit ALK with mutations L1196M and G1269A, respectively. This computational investigation in-sighted the molecular factors involved in crizotinib resistance which enhanced in the identification of new ALK drugs that brings individualized medicine to treat ALK positive NSCLC patients with specific mutations. J. Cell. Biochem. 118: 3462-3471, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nagarajan Nagasundaram
- Molecular and Nanomedicne Research Unit, Centre for Nanoscience and Nanotechnology, Sathyabama University, Jeppiaar Nagar, Chennai 600119, Tamil Nadu, India
| | - Carlton Ranjith Wilson Alphonse
- Molecular and Nanomedicne Research Unit, Centre for Nanoscience and Nanotechnology, Sathyabama University, Jeppiaar Nagar, Chennai 600119, Tamil Nadu, India
| | - Prakash Vincent Samuel Gnana
- Centre for Marine Science and Technology (CMST), Manonmaniam Sundaranar University, Rajakkamangalam, Kanyakumari District 629502, Tamil Nadu, India
| | - Rajesh Kannan Rajaretinam
- Molecular and Nanomedicne Research Unit, Centre for Nanoscience and Nanotechnology, Sathyabama University, Jeppiaar Nagar, Chennai 600119, Tamil Nadu, India
| |
Collapse
|
300
|
Chacko S, Samanta S. A novel approach towards design, synthesis and evaluation of some Schiff base analogues of 2-aminopyridine and 2-aminobezothiazole against hepatocellular carcinoma. Biomed Pharmacother 2017; 89:162-176. [DOI: 10.1016/j.biopha.2017.01.108] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/08/2017] [Accepted: 01/17/2017] [Indexed: 02/07/2023] Open
|