1
|
Li L, Dai S, Liu JY, Wu W, Zhao QX, Wang X, Wang N, Xu ZH. Antagonistic Effect and In Vitro Activity of Dauricine on Glucagon Receptor. JOURNAL OF NATURAL PRODUCTS 2022; 85:2035-2043. [PMID: 35834753 DOI: 10.1021/acs.jnatprod.2c00446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Abnormal increases in glucagon (GCG) are the primary cause of type II diabetes mellitus. When GCG interacts with a glucagon receptor (GCGR), GCG can increase the blood glucose level. In this paper, a compound that could interfere with the binding of GCG and GCGR to inhibit the increase of blood glucose was investigated. First, molecular docking was used to conduct preliminary screening of compounds whose active components could combine with GCGR by AutoDock Vina. The binding of the receptor-ligand complex was analyzed by PyMOL. Results showed that dauricine could tightly bind to the receptor pocket. Second, the plasmid pcDNA3.1(+)-GCGR containing the target gene was transfected into HEK293 cells for expression, which was the cell model established to screen GCGR antagonist. Dauricine, the lead compound of glucagon receptor antagonist (GRA), was screened using the GRA screening model in vitro. Finally, using [Des-His1, Glu9]-Glucagon amide as the positive control, flow cytometry was used to express the antagonistic effect of the compound. Consequently, dauricine can antagonize the GCGR.
Collapse
Affiliation(s)
- Li Li
- College of Science, Xihua University, Chengdu 610039, China
| | - Shuang Dai
- College of Science, Xihua University, Chengdu 610039, China
| | - Jing-Ya Liu
- College of Science, Xihua University, Chengdu 610039, China
| | - Wei Wu
- College of Science, Xihua University, Chengdu 610039, China
| | - Qian-Xi Zhao
- College of Science, Xihua University, Chengdu 610039, China
| | - Xin Wang
- College of Science, Xihua University, Chengdu 610039, China
| | - Na Wang
- College of Science, Xihua University, Chengdu 610039, China
| | - Zhi-Hong Xu
- College of Science, Xihua University, Chengdu 610039, China
| |
Collapse
|
2
|
Identify promising IKK-β inhibitors: A docking-based 3D-QSAR study combining molecular design and molecular dynamics simulation. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.103786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
3
|
Mahnam K, Shakhsi-Niaei M, Ziaei M, Sweazea KL. In silico evaluation of the downstream effect of mutated glucagon is consistent with higher blood glucose homeostasis in Galliformes and Strigiformes. Gen Comp Endocrinol 2021; 314:113925. [PMID: 34624309 DOI: 10.1016/j.ygcen.2021.113925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 11/29/2022]
Abstract
In contrast to mammals, glucagon is reported as a much more potent blood glucose modulator in birds. Interestingly, we have found p.Thr16Ser mutation, a variation in the highly conserved glucagon hormone, in Galliformes as well as Strigiformes. To check the effect of this mutation on the receptor binding of glucagon, we predicted the ancestral glucagon receptor sequence of all available Galliformes and Strigiformes species. Subsequently, we analysed their binding to the mutated and wild type glucagon (ancestral) by molecular dynamics simulation. At first, we made a model of ancestral glucagon receptor and ancestral mutated, and wild type glucagon in the order Galliformes and Strigiformes. Then we performed molecular dynamics for each Galliformes and Strigiformes receptor as well as each glucagon peptide, respectively. The final structures were used for docking simulation of glucagon to their receptors. The results of the docking simulations showed a stronger binding affinity of mutated glucagon to glucagon receptors. Afterward, we obtained blood glucose concentrations of all available Galliformes members, as well as all available members of its only taxonomic neighbour (order Anseriformes) in superorder Galloanserae. Interestingly the p.Thr16Ser mutation could finely cluster these two orders into two groups: higher blood glucose concentration (order Galliformes, 17.64 ± 1.66 mMol/L) and lower blood glucose concentration (order Anseriformes, 11.34 ± 1.11 mMol/L). Strigiformes which carry the mutated glucagon peptide show also high blood glucose concentrations (17.40 ± 1.51 mMol/L). Therefore, the results suggest this mutation, which leads to stronger binding affinity of mutated glucagon to its receptor, may be a driving force for higher blood glucose homeostasis in the related birds.
Collapse
Affiliation(s)
- Karim Mahnam
- Department of Biology, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Mostafa Shakhsi-Niaei
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran.
| | - Maryam Ziaei
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | | |
Collapse
|
4
|
Chen W, Li A, Wang J, Zhong H, Yuan J, Luo Y, Ou J, Chen J, Li L. A Combined approach of QSAR study, molecular docking and pharmacokinetics prediction of promising Amide-Ac6-aminoacetonitriles Cathepsin K inhibitors. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
5
|
Peters C, Bascuñán D, Burgos CF, Bobadilla C, González-Sanmiguel J, Boopathi S, Riffo N, Fernández-Pérez EJ, Tarnok ME, Aguilar LF, Gonzalez W, Aguayo LG. Characterization of a new molecule capable of inhibiting several steps of the amyloid cascade in Alzheimer's disease. Neurobiol Dis 2020; 141:104938. [PMID: 32434047 DOI: 10.1016/j.nbd.2020.104938] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/04/2020] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder in elderly people. Existent therapies are directed at alleviating some symptoms, but are not effective in altering the course of the disease. METHODS Based on our previous study that showed that an Aβ-interacting small peptide protected against the toxic effects of amyloid-beta peptide (Aβ), we carried out an array of in silico, in vitro, and in vivo assays to identify a molecule having neuroprotective properties. RESULTS In silico studies showed that the molecule, referred to as M30 (2-Octahydroisoquinolin-2(1H)-ylethanamine), was able to interact with the Aβ peptide. Additionally, in vitro assays showed that M30 blocked Aβ aggregation, association to the plasma membrane, synaptotoxicity, intracellular calcium, and cellular toxicity, while in vivo experiments demonstrated that M30 induced a neuroprotective effect by decreasing the toxicity of Aβ in the dentate gyrus of the hippocampus and improving the alteration in spatial memory in behavior assays. DISCUSSION Therefore, we propose that this new small molecule could be a useful candidate for the additional development of a treatment against AD since it appears to block multiple steps in the amyloid cascade. Overall, since there are no drugs that effectively block the progression of AD, this approach represents an innovative strategy. SIGNIFICANCE Currently, there is no effective treatment for AD and the expectations to develop an effective therapy are low. Using in silico, in vitro, and in vivo experiments, we identified a new compound that is able to inhibit Aβ-induced neurotoxicity, specifically aggregation, association to neurons, synaptic toxicity, calcium dyshomeostasis and memory impairment induced by Aβ. Because Aβ toxicity is central to AD progression, the inhibition mediated by this new molecule might be useful as a therapeutic tool.
Collapse
Affiliation(s)
- Christian Peters
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepción, Chile
| | - Denisse Bascuñán
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepción, Chile
| | - Carlos F Burgos
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepción, Chile
| | - Catalina Bobadilla
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepción, Chile
| | | | - Subramanian Boopathi
- The Center for Bioinformatics and Molecular Simulations (CBSM), Universidad de Talca, Talca, Chile
| | - Nicolás Riffo
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepción, Chile
| | - Eduardo J Fernández-Pérez
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepción, Chile
| | - María Elena Tarnok
- Laboratory of Photophysics and Molecular Spectroscopy, Chemistry, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Luis Felipe Aguilar
- Laboratory of Photophysics and Molecular Spectroscopy, Chemistry, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Wendy Gonzalez
- The Center for Bioinformatics and Molecular Simulations (CBSM), Universidad de Talca, Talca, Chile; Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Talca, Talca, Chile
| | - Luis G Aguayo
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
6
|
Zhu L, Dattaroy D, Pham J, Wang L, Barella LF, Cui Y, Wilkins KJ, Roth BL, Hochgeschwender U, Matschinsky FM, Kaestner KH, Doliba NM, Wess J. Intra-islet glucagon signaling is critical for maintaining glucose homeostasis. JCI Insight 2019; 5:127994. [PMID: 31012868 DOI: 10.1172/jci.insight.127994] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Glucagon, a hormone released from pancreatic alpha-cells, plays a key role in maintaining proper glucose homeostasis and has been implicated in the pathophysiology of diabetes. In vitro studies suggest that intra-islet glucagon can modulate the function of pancreatic beta-cells. However, because of the lack of suitable experimental tools, the in vivo physiological role of this intra-islet cross-talk has remained elusive. To address this issue, we generated a novel mouse model that selectively expressed an inhibitory designer G protein-coupled receptor (Gi DREADD) in α-cells only. Drug-induced activation of this inhibitory designer receptor almost completely shut off glucagon secretion in vivo, resulting in significantly impaired insulin secretion, hyperglycemia, and glucose intolerance. Additional studies with mouse and human islets indicated that intra-islet glucagon stimulates insulin release primarily by activating β-cell GLP-1 receptors. These new findings strongly suggest that intra-islet glucagon signaling is essential for maintaining proper glucose homeostasis in vivo. Our work may pave the way toward the development of novel classes of antidiabetic drugs that act by modulating intra-islet cross-talk between α- and β-cells.
Collapse
Affiliation(s)
- Lu Zhu
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Diptadip Dattaroy
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Jonathan Pham
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Lingdi Wang
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung and Blood Institute, Bethesda, Maryland, USA
| | - Luiz F Barella
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Yinghong Cui
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Kenneth J Wilkins
- Biostatistics Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Bryan L Roth
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Ute Hochgeschwender
- Neuroscience Program and College of Medicine, Central Michigan University, Mt. Pleasant, Michigan, USA
| | - Franz M Matschinsky
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Klaus H Kaestner
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nicolai M Doliba
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| |
Collapse
|
7
|
Xie X, Liu K, Liu F, Chen H, Wang X, Zu X, Ma X, Wang T, Wu Q, Zheng Y, Bode AM, Dong Z, Kim DJ. Gossypetin is a novel MKK3 and MKK6 inhibitor that suppresses esophageal cancer growth in vitro and in vivo. Cancer Lett 2018; 442:126-136. [PMID: 30391783 DOI: 10.1016/j.canlet.2018.10.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 10/11/2018] [Accepted: 10/22/2018] [Indexed: 01/04/2023]
Abstract
Gossypetin as a hexahydroxylated flavonoid found in many flowers and Hibiscus. It exerts various pharmacological activities, including antioxidant, antibacterial and anticancer activities. However, the anticancer capacity of gossypetin has not been fully elucidated. In this study, gossypetin was found to inhibit anchorage-dependent and -independent growth of esophageal cancer cells. To identify the molecular target(s) of gossypetin, various signaling protein kinases were screened and results indicate that gossypetin strongly attenuates the MKK3/6-p38 signaling pathway by directly inhibiting MKK3 and MKK6 protein kinase activity in vitro. Mechanistic investigations showed that arginine-61 in MKK6 is critical for binding with gossypetin. Additionally, the inhibition of cell growth by gossypetin is dependent on the expression of MKK3 and MKK6. Gossypetin caused G2 phase cell cycle arrest and induced intrinsic apoptosis by activating caspases 3 and 7 and increasing the expression of BAX and cytochrome c. Notably, gossypetin suppressed patient-derived esophageal xenograft tumor growth in an in vivo mouse model. Our findings suggest that gossypetin is an MKK3 and MKK6 inhibitor that could be useful for preventing or treating esophageal cancer.
Collapse
Affiliation(s)
- Xiaomeng Xie
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450008, China
| | - Kangdong Liu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450008, China; The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450008, China; The Affiliated Cancer Hospital, Zhengzhou University, Zhengzhou, Henan, 450008, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450008, China
| | - Feifei Liu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450008, China
| | - Hanyong Chen
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Xiangyu Wang
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450008, China
| | - Xueyin Zu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450008, China; The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Xiaoli Ma
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450008, China; The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Ting Wang
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450008, China; The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Qiong Wu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450008, China; The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Yan Zheng
- The Affiliated Cancer Hospital, Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Zigang Dong
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450008, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450008, China; The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA.
| | - Dong Joon Kim
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450008, China.
| |
Collapse
|
8
|
Goswami D, Goyal S, Jamal S, Jain R, Wahi D, Grover A. GQSAR modeling and combinatorial library generation of 4-phenylquinazoline-2-carboxamide derivatives as antiproliferative agents in human Glioblastoma tumors. Comput Biol Chem 2017; 69:147-152. [PMID: 28420545 DOI: 10.1016/j.compbiolchem.2017.03.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 03/27/2017] [Indexed: 12/17/2022]
|
9
|
Dholakia D, Goyal S, Jamal S, Singh A, Das A, Grover A. Molecular modeling and lead design of substituted zanamivir derivatives as potent anti-influenza drugs. BMC Bioinformatics 2016; 17:512. [PMID: 28155702 PMCID: PMC5259988 DOI: 10.1186/s12859-016-1374-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Influenza virus spreads infection by two main surface glycoproteins, namely hemagglutinin (HA) and neuraminidase (NA). NA cleaves the sialic acid receptors eventually releasing newly formed virus particles which then invade new cells. Inhibition of NA could limit the replication of virus to one round which is insufficient to cause the disease. RESULTS An experimentally reported series of acylguanidine zanamivir derivatives was used to develop GQSAR model targeting NA in different strains of influenza virus, H1N1 and H3N2. A combinatorial library was developed and their inhibitory activities were predicted using the GQSAR model. CONCLUSION The top leads were analyzed by docking which revealed the binding modes of these inhibitors in the active site of NA (150-loop). The top compound (AMA) was selected for carrying out molecular dynamics simulations for 15 ns which provided insights into the time dependent dynamics of the designed leads. AMA possessed a docking score of -8.26 Kcal/mol with H1N1 strain and -7.00 Kcal/mol with H3N2 strain. Ligand-bound complexes of both H1N1 and H3N2 were observed to be stable for 11 ns and 7 ns respectively. ADME descriptors were also calculated to study the pharmacokinetic properties of AMA which revealed its drug-like properties.
Collapse
Affiliation(s)
- Dhwani Dholakia
- Department of Biotechnology, Delhi Technological University, New Delhi, 110042 India
| | - Sukriti Goyal
- Department of Bioscience and Biotechnology, Banasthali University, Tonk, Rajasthan 304022 India
| | - Salma Jamal
- Department of Bioscience and Biotechnology, Banasthali University, Tonk, Rajasthan 304022 India
| | - Aditi Singh
- Department of Biotechnology, TERI University, New Delhi, 110070 India
| | - Asmita Das
- Department of Biotechnology, Delhi Technological University, New Delhi, 110042 India
| | - Abhinav Grover
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067 India
| |
Collapse
|
10
|
Nagpal N, Goyal S, Dhanjal JK, Ye L, Kaul SC, Wadhwa R, Chaturvedi R, Grover A. Molecular dynamics-based identification of novel natural mortalin-p53 abrogators as anticancer agents. J Recept Signal Transduct Res 2016; 37:8-16. [PMID: 27380217 DOI: 10.3109/10799893.2016.1141952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Cancer is one of the leading causes of mortality worldwide that requires attention in terms of extensive study and research. Eradication of mortalin-p53 interaction that leads to the inhibition of transcriptional activation or blocking of p53 from functioning as a suppressor and induction of nuclear translocation of p53 can prove to be one of the useful approaches for cancer management. RESULTS In this study, we used structure-based approach to target the p53-binding domain of mortalin in order to prevent mortalin-p53 complex formation. We screened compounds from ZINC database against the modeled mortalin protein using Glide virtual screening. The top two compounds, DTOM (ZINC 28639308) and TTOM (ZINC 38143676) with Glide score of -12.27 and -12.16, respectively, were identified with the potential to abrogate mortalin-p53 interaction. Finally, molecular dynamics simulations were used to analyze the dynamic stability of the ligand-bound complex and it was observed that residues Tyr196, Asn198, Val264 and Thr267 were involved in intermolecular interactions in both the simulated ligand-bound complexes, and thus, these residues may have a paramount role in stabilizing the binding of the ligands with the protein. CONCLUSION These detailed insights can further facilitate the development of potent inhibitors against mortalin-p53 complex.
Collapse
Affiliation(s)
- Neha Nagpal
- a School of Biotechnology, Jawaharlal Nehru University , New Delhi , India and
| | - Sukriti Goyal
- a School of Biotechnology, Jawaharlal Nehru University , New Delhi , India and
| | | | - Liu Ye
- b Cell Proliferation Research Group and DBT-AIST International Laboratory for Advanced Biomedicine, National Institute of Advanced Industrial Science & Technology (AIST) , Tsukuba , Ibaraki , Japan
| | - Sunil C Kaul
- b Cell Proliferation Research Group and DBT-AIST International Laboratory for Advanced Biomedicine, National Institute of Advanced Industrial Science & Technology (AIST) , Tsukuba , Ibaraki , Japan
| | - Renu Wadhwa
- b Cell Proliferation Research Group and DBT-AIST International Laboratory for Advanced Biomedicine, National Institute of Advanced Industrial Science & Technology (AIST) , Tsukuba , Ibaraki , Japan
| | - Rupesh Chaturvedi
- a School of Biotechnology, Jawaharlal Nehru University , New Delhi , India and
| | - Abhinav Grover
- a School of Biotechnology, Jawaharlal Nehru University , New Delhi , India and
| |
Collapse
|
11
|
Singh A, Goyal S, Jamal S, Subramani B, Das M, Admane N, Grover A. Computational identification of novel piperidine derivatives as potential HDM2 inhibitors designed by fragment-based QSAR, molecular docking and molecular dynamics simulations. Struct Chem 2016; 27:993-1003. [DOI: 10.1007/s11224-015-0697-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
12
|
Interaction of Glucagon G-Protein Coupled Receptor with Known Natural Antidiabetic Compounds: Multiscoring In Silico Approach. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:497253. [PMID: 26236379 PMCID: PMC4508340 DOI: 10.1155/2015/497253] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 06/15/2015] [Indexed: 01/18/2023]
Abstract
Glucagon receptor (GCGR) is a secretin-like (class B) family of G-protein coupled receptors (GPCRs) in humans that plays an important role in elevating the glucose concentration in blood and has thus become one of the promising therapeutic targets for treatment of type 2 diabetes mellitus. GCGR based inhibitors for the treatment of type 2 diabetes are either glucagon neutralizers or small molecular antagonists. Management of diabetes without any side effects is still a challenge to the medical system, and the search for a new and effective natural GCGR antagonist is an important area for the treatment of type 2 diabetes. In the present study, a number of natural compounds containing antidiabetic properties were selected from the literature and their binding potential against GCGR was determined using molecular docking and other in silico approaches. Among all selected natural compounds, curcumin was found to be the most effective compound against GCGR followed by amorfrutin 1 and 4-hydroxyderricin. These compounds were rescored to confirm the accuracy of binding using another scoring function (x-score). The final conclusions were drawn based on the results obtained from the GOLD and x-score. Further experiments were conducted to identify the atomic level interactions of selected compounds with GCGR.
Collapse
|
13
|
Nagpal N, Goyal S, Wahi D, Jain R, Jamal S, Singh A, Rana P, Grover A. Molecular principles behind Boceprevir resistance due to mutations in hepatitis C NS3/4A protease. Gene 2015; 570:115-21. [PMID: 26055089 DOI: 10.1016/j.gene.2015.06.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/28/2015] [Accepted: 06/03/2015] [Indexed: 02/05/2023]
Abstract
The hepatitis C virus (HCV) infection is a primary cause of chronic hepatitis which eventually progresses to cirrhosis and in some instances might advance to hepatocellular carcinoma. According to the WHO report, HCV infects 130-150 million people globally and every year 350,000 to 500,000 people die from hepatitis C virus infection. Great achievement has been made in viral treatment evolution, after the development of HCV NS3/4A protease inhibitor (Boceprevir). However, efficacy of Boceprevir is compromised by the emergence of drug resistant variants. The molecular principle behind drug resistance of the protease mutants such as (V36M, T54S and R155K) is still poorly understood. Therefore in this study, we employed a series of computational strategies to analyze the binding of antiviral drug, Boceprevir to HCV NS3/4A protease mutants. Our results clearly demonstrate that the point mutations (V36M, T54S and R155K) in protease are associated with lowering of its binding affinity with Boceprevir. Exhaustive analysis of the simulated Boceprevir-bound wild and mutant complexes revealed variations in hydrophobic interactions, hydrogen bond occupancy and salt bridge interactions. Also, substrate envelope analysis scrutinized that the studied mutations reside outside the substrate envelope which may affect the Boceprevir affinity towards HCV protease but not the protease enzymatic activity. Furthermore, structural analyses of the binding site volume and flexibility show impairment in flexibility and stability of the binding site residues in mutant structures. In order to combat Boceprevir resistance, renovation of binding interactions between the drug and protease may be valuable. The structural insight from this study reveals the mechanism of the Boceprevir resistance and the results can be valuable for the design of new PIs with improved efficiency.
Collapse
Affiliation(s)
- Neha Nagpal
- Department of Biotechnology, Delhi Technological University, Delhi 110042, India
| | - Sukriti Goyal
- Department of Bioscience and Biotechnology, Banasthali University, Tonk, Rajasthan 304022, India
| | - Divya Wahi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Ritu Jain
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Salma Jamal
- Department of Bioscience and Biotechnology, Banasthali University, Tonk, Rajasthan 304022, India
| | - Aditi Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Preeti Rana
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Abhinav Grover
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
14
|
Deng D, Li Y, Xue J, Wang J, Ai G, Li X, Gu Y. Gold nanoparticle-based beacon to detect STAT5b mRNA expression in living cells: a case optimized by bioinformatics screen. Int J Nanomedicine 2015; 10:3231-44. [PMID: 25987838 PMCID: PMC4422291 DOI: 10.2147/ijn.s81754] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Messenger RNA (mRNA), a single-strand ribonucleic acid with functional gene information is usually abnormally expressed in cancer cells and has become a promising biomarker for the study of tumor progress. Hairpin DNA-coated gold nanoparticle (hDAuNP) beacon containing a bare gold nanoparticle (AuNP) as fluorescence quencher and thiol-terminated fluorescently labeled stem-loop-stem oligonucleotide sequences attached by Au-S bond is currently a new nanoscale biodiagnostic platform capable of mRNA detection, in which the design of the loop region sequence is crucial for hybridizing with the target mRNA. Hence, in this study, to improve the sensitivity and selectivity of hDAuNP beacon simultaneously, the loop region of hairpin DNA was screened by bioinformatics strategy. Here, signal transducer and activator of transcription 5b (STAT5b) mRNA was selected and used as a practical example. The results from the combined characterizations using optical techniques, flow cytometry assay, and cell microscopic imaging showed that after optimization, the as-prepared hDAuNP beacon had higher selectivity and sensitivity for the detection of STAT5b mRNA in living cells, as compared with our previous beacon. Thus, the bioinformatics method may be a promising new strategy for assisting in the designing of the hDAuNP beacon, extending its application in the detection of mRNA expression and the resultant mRNA-based biological processes and disease pathogenesis.
Collapse
Affiliation(s)
- Dawei Deng
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yang Li
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jianpeng Xue
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jie Wang
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Guanhua Ai
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xin Li
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yueqing Gu
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
15
|
Filipski KJ. Small molecule glucagon receptor antagonists: a patent review (2011 – 2014). Expert Opin Ther Pat 2015; 25:819-30. [DOI: 10.1517/13543776.2015.1032250] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
16
|
Ranganathan S, Tan TW, Schönbach C. InCoB2014: bioinformatics to tackle the data to knowledge challenge. Introduction. BMC Bioinformatics 2014; 15 Suppl 16:I1. [PMID: 25521055 PMCID: PMC4290632 DOI: 10.1186/1471-2105-15-s16-i1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Since 2006, the International Conference on Bioinformatics (InCoB) has been publishing selected papers in BMC Bioinformatics. Papers within the scope of the journal from the 13th InCoB July 31-2 August, 2014 in Sydney, Australia have been compiled in this supplement. These span protein and proteome informatics, structural bioinformatics, software development and bioimaging to pharmacoinformatics and disease informatics, representing the breadth of bioinformatics research in the Asia-Pacific.
Collapse
Affiliation(s)
- Shoba Ranganathan
- Department of Chemistry and Biomolecular Sciences and ARC Centre of Excellence in Bioinformatics, Macquarie University, Sydney NSW 2109, Australia
| | - Tin Wee Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599
| | - Christian Schönbach
- Department of Biology, School of Science and Technology, Nazarbayev University, Astana 010000, Republic of Kazakhstan
- Center for AIDS Research, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|