1
|
Sivadas A, Rathore S, Sahana S, Jolly B, Bhoyar RC, Jain A, Sharma D, Imran M, Senthilvel V, Divakar MK, Mishra A, Sivasubbu S, Scaria V. The genomic landscape of CYP2D6 variation in the Indian population. Pharmacogenomics 2024; 25:147-160. [PMID: 38426301 DOI: 10.2217/pgs-2023-0233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Abstract
Aim: The CYP2D6 gene is highly polymorphic, causing large interindividual variability in the metabolism of several clinically important drugs. Materials & methods: The authors investigated the diversity and distribution of CYP2D6 alleles in Indians using whole genome sequences (N = 1518). Functional consequences were assessed using pathogenicity scores and molecular dynamics simulations. Results: The analysis revealed population-specific CYP2D6 alleles (*86, *7, *111, *112, *113, *99) and remarkable differences in variant and phenotype frequencies with global populations. The authors observed that one in three Indians could benefit from a dose alteration for psychiatric drugs with accurate CYP2D6 phenotyping. Molecular dynamics simulations revealed large conformational fluctuations, confirming the predicted reduced function of *86 and *113 alleles. Conclusion: The findings emphasize the utility of comprehensive CYP2D6 profiling for aiding precision public health.
Collapse
Affiliation(s)
- Ambily Sivadas
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, Karnataka, 560034, India
| | - Surabhi Rathore
- CSIR Institute of Genomics & Integrative Biology, Mathura Road, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - S Sahana
- CSIR Institute of Genomics & Integrative Biology, Mathura Road, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Bani Jolly
- CSIR Institute of Genomics & Integrative Biology, Mathura Road, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Rahul C Bhoyar
- CSIR Institute of Genomics & Integrative Biology, Mathura Road, New Delhi, 110025, India
| | - Abhinav Jain
- CSIR Institute of Genomics & Integrative Biology, Mathura Road, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Disha Sharma
- CSIR Institute of Genomics & Integrative Biology, Mathura Road, New Delhi, 110025, India
| | - Mohamed Imran
- CSIR Institute of Genomics & Integrative Biology, Mathura Road, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Vigneshwar Senthilvel
- CSIR Institute of Genomics & Integrative Biology, Mathura Road, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Mohit Kumar Divakar
- CSIR Institute of Genomics & Integrative Biology, Mathura Road, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Anushree Mishra
- CSIR Institute of Genomics & Integrative Biology, Mathura Road, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Sridhar Sivasubbu
- CSIR Institute of Genomics & Integrative Biology, Mathura Road, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
- Vishwanath Cancer Care Foundation, B 702, 7th Floor, Neelkanth Business Park Kirol Village, Vidya Vihar, West Mumbai, 400086, India
| | - Vinod Scaria
- CSIR Institute of Genomics & Integrative Biology, Mathura Road, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
- Vishwanath Cancer Care Foundation, B 702, 7th Floor, Neelkanth Business Park Kirol Village, Vidya Vihar, West Mumbai, 400086, India
| |
Collapse
|
2
|
Niwa T. [Metabolic Activities Catalyzed by Human Cytochrome P450 (CYP) 2D6 and CYP3A Subfamily Members and Effect of Various Compounds, Including Endogenous Steroid Hormones, on These Activities]. YAKUGAKU ZASSHI 2024; 144:197-202. [PMID: 38296497 DOI: 10.1248/yakushi.23-00174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
My research focused on the effects of various drugs on (1) dopamine formation from p-tyramine catalyzed by polymorphic cytochrome P450 (CYP or P450) 2D6 variants and (2) endogenous steroid hormone hydroxylation catalyzed by CYP3A subfamily members (CYP3A4, CYP3A5, CYP3A7). The activation (cooperativity) of metabolic reactions catalyzed by P450s was especially emphasized. The effects of various psychotropic agents on dopamine formation from p-tyramine, catalyzed by wild-type CYP2D6.1 and CYP2D6 variants, including CYP2D6.2 (Arg296Cys;Ser486Thr), CYP2D6.10 (Pro34Ser;Ser486Thr), and CYP2D6.39 (Ser486Thr) were compared. Michaelis (Km) and inhibition (Ki) constants of the psychotropic agents in the presence of CYP2D6.10 were higher than those observed in the presence of other CYP2D6 variants. Fluvoxamine, fluoxetine, milnacipran, and haloperidol activated CYP2D6-catalyzed dopamine formation [decreasing the Km and/or increasing the maximal velocity (kcat)], and this activation was CYP2D6 variant-dependent. Regarding the CYP3A subfamily, the effects of various compounds including endogenous steroid hormones on the 6β-hydroxylation of steroid hormones, such as testosterone, progesterone, and cortisol, were determined; it was found that testosterone, dehydroepiandrosterone, and/or α-naphthoflavone activated 6β-hydroxylation of cortisol and/or progesterone, but the effects varied in the presence of different CYP3A subfamily members. Further studies are required to confirm the mechanisms and therapeutic relevance of these activation phenomena.
Collapse
|
3
|
Dong AN, Ahemad N, Pan Y, Palanisamy UD, Yiap BC, Ong CE. Role of P34S, G169R, R296C, and S486T Substitutions in Ligand Access and Catalysis for Cytochrome P450 2D6 Allelic Variants CYP2D6*14A and CYP2D6*14B. DRUG METABOLISM AND BIOANALYSIS LETTERS 2022; 15:51-63. [PMID: 35049443 DOI: 10.2174/1872312815666220113125232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/16/2021] [Accepted: 12/17/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Genetic polymorphism of cytochrome P450 (CYP) contributes to variability in drug metabolism, clearance, and response. This study aimed to investigate the functional and molecular basis for altered ligand binding and catalysis in CYP2D6*14A and CYP2D6*14B, two unique alleles common in the Asian population. METHODS CYP proteins expressed in Escherichia coli were studied using the substrate 3-cyano-7- ethoxycoumarin (CEC) and inhibitor probes (quinidine, fluoxetine, paroxetine, terbinafine) in the enzyme assay. Computer modelling was additionally used to create three-dimensional structures of the CYP2D6*14 variants. RESULTS Kinetics data indicated significantly reduced intrinsic clearance in CYP2D6*14 variants, suggesting that P34S, G169R, R296C, and S486T substitutions worked cooperatively to alter the conformation of the active site that negatively impacted the deethylase activity of CYP2D6. For the inhibition studies, IC50 values decreased in quinidine, paroxetine, and terbinafine but increased in fluoxetine, suggesting a varied ligand-specific susceptibility to inhibition. Molecular docking further demonstrated the role of P34S and R296C in altering access channel dimensions, thereby affecting ligand access and binding and subsequently resulting in varied inhibition potencies. CONCLUSION In summary, the differential selectivity of CYP2D6*14 variants for the ligands (substrate and inhibitor) was governed by the alteration of the active site and access channel architecture induced by the natural mutations found in the alleles.
Collapse
Affiliation(s)
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Selangor, Malaysia
| | - Yan Pan
- Department of Biomedical Science, University of Nottingham, Malaysia Campus, Semenyih, Selangor, Malaysia
| | - Uma Devi Palanisamy
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Selangor, Malaysia
| | - Beow Chin Yiap
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Chin Eng Ong
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
A Conservative Replacement in the Transmembrane Domain of SARS-CoV-2 ORF7a as a Putative Risk Factor in COVID-19. BIOLOGY 2021; 10:biology10121276. [PMID: 34943191 PMCID: PMC8698902 DOI: 10.3390/biology10121276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/26/2021] [Accepted: 12/03/2021] [Indexed: 01/02/2023]
Abstract
Simple Summary The pathogenicity and transmissibility of the COVID-19 pandemic causative agent, the SARS-CoV-2 virus, is related to the functions of the proteins synthesized intracellularly, as guided by viral RNA. It is vitally important to accurately pinpoint novel variants of concern of the SARS-CoV-2 virus, in order to understand the molecular features of novel mutations and manage the on-going battle against the COVID-19 pandemic. We focused on A105V mutation in the ORF7a accessory protein. Sequencing and clinical data showed that this mutation is associated with increased severity and lethality in a group of Romanian patients, despite a lower viral copy number and a lower number of associated comorbidities. This effect is primarily due to increased protein stability through allosteric effects as shown by molecular dynamics analyses. This behavior manifests especially among residues 39–56, and the ones adjacent to 26–30 loop, placed in direct contact with potential interaction partners. Together, the results provide novel insights into the role of ORF7a in the pathogenicity of SARS-CoV-2. Abstract The ongoing COVID-19 pandemic follows an unpredictable evolution, driven by both host-related factors such as mobility, vaccination status, and comorbidities and by pathogen-related ones. The pathogenicity of its causative agent, SARS-CoV-2 virus, relates to the functions of the proteins synthesized intracellularly, as guided by viral RNA. These functions are constantly altered through mutations resulting in increased virulence, infectivity, and antibody-evasion abilities. Well-characterized mutations in the spike protein, such as D614G, N439K, Δ69–70, E484K, or N501Y, are currently defining specific variants; however, some less studied mutations outside the spike region, such as p. 3691 in NSP6, p. 9659 in ORF-10, 8782C > T in ORF-1ab, or 28144T > C in ORF-8, have been proposed for altering SARS-CoV-2 virulence and pathogenicity. Therefore, in this study, we focused on A105V mutation of SARS-CoV-2 ORF7a accessory protein, which has been associated with severe COVID-19 clinical manifestation. Molecular dynamics and computational structural analyses revealed that this mutation differentially alters ORF7a dynamics, suggesting a gain-of-function role that may explain its role in the severe form of COVID-19 disease.
Collapse
|
5
|
Mohanty L, Mishra C, Pradhan SK, Mishra SR, Nayak G. Identification of novel polymorphism and in silico analysis of caprine DNAJB3 gene. Small Rumin Res 2021. [DOI: 10.1016/j.smallrumres.2021.106492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
6
|
Deciphering Structural Alterations Associated with Activity Reductions of Genetic Polymorphisms in Cytochrome P450 2A6 Using Molecular Dynamics Simulations. Int J Mol Sci 2021; 22:ijms221810119. [PMID: 34576282 PMCID: PMC8469730 DOI: 10.3390/ijms221810119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/04/2022] Open
Abstract
Cytochrome P450 (CYP) 2A6 is a monooxygenase involved in the metabolism of various endogenous and exogenous chemicals, such as nicotine and therapeutic drugs. The genetic polymorphisms in CYP2A6 are a cause of individual variation in smoking behavior and drug toxicities. The enzymatic activities of the allelic variants of CYP2A6 were analyzed in previous studies. However, the three-dimensional structures of the mutants were not investigated, and the mechanisms underlying activity reduction remain unknown. In this study, to investigate the structural changes involved in the reduction in enzymatic activities, we performed molecular dynamics simulations for ten allelic mutants of CYP2A6. For the calculated wild type structure, no significant structural changes were observed in comparison with the experimental structure. On the other hand, the mutations affected the interaction with heme, substrates, and the redox partner. In CYP2A6.44, a structural change in the substrate access channel was also observed. Those structural effects could explain the alteration of enzymatic activity caused by the mutations. The results of simulations provide useful information regarding the relationship between genotype and phenotype.
Collapse
|
7
|
Virtual Alanine Scan of the Main Protease Active Site in Severe Acute Respiratory Syndrome Coronavirus 2. Int J Mol Sci 2021; 22:ijms22189837. [PMID: 34576002 PMCID: PMC8466562 DOI: 10.3390/ijms22189837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 08/29/2021] [Accepted: 09/08/2021] [Indexed: 11/17/2022] Open
Abstract
Recently, inhibitors of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease (Mpro) have been proposed as potential therapeutic agents for COVID-19. Studying effects of amino acid mutations in the conformation of drug targets is necessary for anticipating drug resistance. In this study, with the structure of the SARS-CoV-2 Mpro complexed with a non-covalent inhibitor, we performed molecular dynamics (MD) simulations to determine the conformation of the complex when single amino acid residue in the active site is mutated. As a model of amino acid mutation, we constructed mutant proteins with one residue in the active site mutated to alanine. This method is called virtual alanine scan. The results of the MD simulations showed that the conformation and configuration of the ligand was changed for mutants H163A and E166A, although the structure of the whole protein and of the catalytic dyad did not change significantly, suggesting that mutations in His163 and Glu166 may be linked to drug resistance.
Collapse
|
8
|
Wilson CJ, Chang M, Karttunen M, Choy WY. KEAP1 Cancer Mutants: A Large-Scale Molecular Dynamics Study of Protein Stability. Int J Mol Sci 2021; 22:5408. [PMID: 34065616 PMCID: PMC8161161 DOI: 10.3390/ijms22105408] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/30/2022] Open
Abstract
We have performed 280 μs of unbiased molecular dynamics (MD) simulations to investigate the effects of 12 different cancer mutations on Kelch-like ECH-associated protein 1 (KEAP1) (G333C, G350S, G364C, G379D, R413L, R415G, A427V, G430C, R470C, R470H, R470S and G476R), one of the frequently mutated proteins in lung cancer. The aim was to provide structural insight into the effects of these mutants, including a new class of ANCHOR (additionally NRF2-complexed hypomorph) mutant variants. Our work provides additional insight into the structural dynamics of mutants that could not be analyzed experimentally, painting a more complete picture of their mutagenic effects. Notably, blade-wise analysis of the Kelch domain points to stability as a possible target of cancer in KEAP1. Interestingly, structural analysis of the R470C ANCHOR mutant, the most prevalent missense mutation in KEAP1, revealed no significant change in structural stability or NRF2 binding site dynamics, possibly indicating an covalent modification as this mutant's mode of action.
Collapse
Affiliation(s)
- Carter J. Wilson
- Department of Biochemistry, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5C1, Canada; (C.J.W.); (M.C.)
- Department of Applied Mathematics, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5B7, Canada
| | - Megan Chang
- Department of Biochemistry, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5C1, Canada; (C.J.W.); (M.C.)
| | - Mikko Karttunen
- Department of Applied Mathematics, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5B7, Canada
- Department of Chemistry, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 3K7, Canada
- Centre for Advanced Materials and Biomaterials Research, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5B7, Canada
| | - Wing-Yiu Choy
- Department of Biochemistry, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5C1, Canada; (C.J.W.); (M.C.)
| |
Collapse
|
9
|
Karn R, Emerson IA. Molecular dynamic study on PTEN frameshift mutations in breast cancer provide c2 domain as a potential biomarker. J Biomol Struct Dyn 2020; 40:3132-3143. [PMID: 33183179 DOI: 10.1080/07391102.2020.1845802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PTEN is a tumour suppressor gene known for regulating apoptosis, cell growth, and many other pathways. It is one of the most frequently mutated genes comprising the phosphatase domain (PD) and C terminal domain (C2). Direct therapeutic methods are not applicable for targeting PTEN because once gets mutated, it needs restoration. For mutant detection and restoration using PTEN mRNA there is a need to explore various mutations taking place in PTEN, identify their particular domains, and study their interactions within the cellular system. Here, we have tried to highlight a few such regions in the mutated PTEN of breast cancer patients. In this study, we have selected the top-most-occurring PTEN mutation in breast cancer and compared them to determine the specific properties of each mutation and its effect on functionality. Molecular dynamic simulation for 50 ns was performed on five structures to compare the structural behaviour of mutated PTEN in the system. Our finding suggests that frameshift mutations are more damaging and affect the c2 domain. Frameshift mutant fs_ACTT is the highest occurring as well as the most damaging mutation in all the compared structures. Docking study shows that substitution mutations D92H and R130Q causes loss of binding ability towards PIP2 in normal PTEN, interfering the dephosphorylation process. Overall, the C2 domain is more frequently mutated, and the amino acid residues in the C2 domain show more fluctuations compared to the other regions. Our study can provide the basis for selecting frequently mutated C2 domain as a potential therapeutic marker.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rohit Karn
- Bioinformatics Programming Lab, Department of Biotechnology, School of Bio Sciences and Technology, VIT, Vellore, India
| | - Isaac Arnold Emerson
- Bioinformatics Programming Lab, Department of Biotechnology, School of Bio Sciences and Technology, VIT, Vellore, India
| |
Collapse
|
10
|
Houndonougbo Y, Pugh B, VanWormer K, April C, Burgis N. Structural dynamics of inosine triphosphate pyrophosphatase (ITPA) protein and two clinically relevant mutants: molecular dynamics simulations. J Biomol Struct Dyn 2020; 39:1236-1247. [PMID: 32129147 DOI: 10.1080/07391102.2020.1727363] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The inosine triphosphate pyrophosphatase (ITPA) protein is responsible for removing noncanonical purine nucleoside triphosphates from intracellular nucleotide pools. Absence of ITPA results in genomic instability and increased levels of inosine in DNA and RNA. The proline to threonine substitution at position 32 (P32T) affects roughly 15% of the global population and can modulate treatment outcomes for cancer, lupus, and hepatitis C patients. The substitution of arginine with cysteine at position 178 (R178C) is extremely uncommon and has only been reported in a small cohort of early infantile encephalopathy patients suggesting that a functional ITPA protein is required for life in humans. Here we present molecular dynamic simulations that describe the structure and dynamics of the wild-type ITPA homodimer and two of its clinically relevant mutants, P32T and R178C. The simulation results indicate that both the P32T and R178C mutations alter the structure and dynamic properties of the protein and provide a possible explanation of the experimentally observed effect of the mutations on ITPA activity. Specifically, the mutations increased the overall flexibility of the protein and changed the dominant collective motions of the top lobe as well as the helix 2 of the lower lobe. Moreover, we have identified key active-site residues that are classified as essential or intermediate for inosine triphosphate (ITP) hydrolyzing activity based on their hydrogen bond occupancy. Here we also present biochemical data indicating that the R178C mutant has very low ITP hydrolyzing activity.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yao Houndonougbo
- Department of Chemistry and Biochemistry, Eastern Washington University, Cheney, WA, USA
| | - Bethany Pugh
- Department of Chemistry and Biochemistry, Eastern Washington University, Cheney, WA, USA
| | - Kandise VanWormer
- Department of Chemistry and Biochemistry, Eastern Washington University, Cheney, WA, USA
| | - Caitlin April
- Department of Chemistry and Biochemistry, Eastern Washington University, Cheney, WA, USA
| | - Nicholas Burgis
- Department of Chemistry and Biochemistry, Eastern Washington University, Cheney, WA, USA
| |
Collapse
|
11
|
Xin J, Yuan M, Peng Y, Wang J. Analysis of the Deleterious Single-Nucleotide Polymorphisms Associated With Antidepressant Efficacy in Major Depressive Disorder. Front Psychiatry 2020; 11:151. [PMID: 32256400 PMCID: PMC7093583 DOI: 10.3389/fpsyt.2020.00151] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/18/2020] [Indexed: 12/26/2022] Open
Abstract
Major depressive disorder (MDD) is a serious mental disease with negative effects on both mental and physical health of the patient. Currently, antidepressants are among the major ways to ease or treat MDD. However, the existing antidepressants have limited efficacy in treating MDD, with a large fraction of patients either responding inadequately or differently to antidepressants during the treatment. Pharmacogenetics studies have found that the genetic features of some genes are associated with the antidepressant efficacy. In order to obtain a better understanding on the relationship between the genetic factors and antidepressant treatment response, we compiled a list of 233 single-nucleotide polymorphisms (SNPs) significantly associated with the antidepressant efficacy in treating MDD. Of the 13 non-synonymous SNPs in the list, three (rs1065852, rs3810651, and rs117986340) may influence the structures and function of the corresponding proteins. Besides, the influence of rs1065852 on the structure of CYP2D6 was further investigated via molecular dynamics simulations. Our results showed that compared to the native CYP2D6 the flexibility of the F-G loop was reduced in the mutant. As a portion of the substrate access channel, the lower flexibility of F-G loop may reduce the ability of the substrates to enter the channel, which may be the reason for the lower enzyme activity of mutant. This study may help us to understand the impact of genetic variation on antidepressant efficacy and provide clues for developing new antidepressants.
Collapse
Affiliation(s)
- Juncai Xin
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Meng Yuan
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Yonglin Peng
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Ju Wang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| |
Collapse
|
12
|
Kolli V, Paul S, Guttula PK, Sarkar N. Elucidating the Role of Val-Asn 95 and Arg-Gly 52 Mutations on Structure and Stability of Fibroblast Growth Factor Homologous Factor 2. Protein Pept Lett 2019; 26:848-859. [PMID: 37020363 DOI: 10.2174/0929866526666190503092718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/11/2019] [Accepted: 04/19/2019] [Indexed: 11/22/2022]
Abstract
Background:
Fibroblast growth Factor Homologous Factors (FHFs) belong to a subclass
of Fibroblast Growth Factor (FGF) family owing to their high sequence and structural similarities
with FGFs. However, despite these similarities, there are properties which set them apart from
FGFs. FHFs lack the secretion signal sequence unlike other FGF members, except FGF1 and 2.
Unlike FGFs, FHFs are not able to bind to FGF Receptors (FGFRs) and instead have been
implicated in binding to Voltage-Gated Sodium Channels (VGSCs), neuronal MAP kinase scaffold
protein and islet-brain-2 (IB2). The two amino acids Arg-52 and Val95 are conserved in all FHFs
and mutation of these residues lead to its inability to bind with VGSC/IB2. However, it is not clear
whether the loss of binding is due to destabilization of the protein on mutation or due to
involvement of Arg52 and Val95 in conferring functionality to FHFs.
Objective:
In the present study, we have mutated these two conserved residues of FHF2 with its
corresponding FGF counterpart amino acids and studied the effects of the mutations on the
structure and stability of the protein.
Methods:
Several biophysical methods like isothermal equilibrium denaturation study, ANS
fluorescence, intrinsic fluorescence, acrylamide quenching, circular dichroism studies as well as
using computational approaches were employed.
Results:
The single mutations were found to affect the overall stability, conformation and
functionality of the protein.
Conclusion:
Thus, the studies throw light on the role of specific amino acids in deciding the
stability, structure and functionality of proteins and will be useful for development of
therapeutically engineered proteins.
Collapse
Affiliation(s)
- Vidyalatha Kolli
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela- 769008, Odisha, India
| | - Subhankar Paul
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela- 769008, Odisha, India
| | - Praveen Kumar Guttula
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela- 769008, Odisha, India
| | - Nandini Sarkar
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela- 769008, Odisha, India
| |
Collapse
|
13
|
Functional and structural characterisation of common cytochrome P450 2D6 allelic variants—roles of Pro34 and Thr107 in catalysis and inhibition. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:1015-1029. [DOI: 10.1007/s00210-019-01651-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 04/09/2019] [Indexed: 02/02/2023]
|
14
|
Ariza Márquez YV, Briceño I, Aristizábal F, Niño LF, Yosa Reyes J. Dynamic Effects of CYP2D6 Genetic Variants in a Set of Poor Metaboliser Patients with Infiltrating Ductal Cancer Under Treatment with Tamoxifen. Sci Rep 2019; 9:2521. [PMID: 30792473 PMCID: PMC6385267 DOI: 10.1038/s41598-018-38340-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/19/2018] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is a group of multigenic diseases. It is the most common cancer diagnosed among women worldwide and is often treated with tamoxifen. Tamoxifen is catalysed by cytochrome P450 2D6 (CYP2D6), and inter-individual variations in the enzyme due to single nucleotide polymorphisms (SNPs) could alter enzyme activity. We evaluated SNPs in patients from Colombia in South America who were receiving tamoxifen treatment for breast cancer. Allelic diversity in the CYP2D6 gene was found in the studied population, with two patients displaying the poor-metaboliser phenotype. Molecular dynamics and trajectory analyses were performed for CYP2D6 from these two patients, comparing it with the common allelic form (CYP2D6*1). Although we found no significant structural change in the protein, its dynamics differ significantly from those of CYP2D6*1, the effect of such differential dynamics resulting in an inefficient enzyme with serious implications for tamoxifen-treated patients, increasing the risk of disease relapse and ineffective treatment.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Agents, Hormonal/administration & dosage
- Antineoplastic Agents, Hormonal/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal/drug therapy
- Carcinoma, Ductal/genetics
- Carcinoma, Ductal/metabolism
- Carcinoma, Ductal/pathology
- Chemotherapy, Adjuvant
- Cytochrome P-450 CYP2D6/genetics
- Cytochrome P-450 CYP2D6/metabolism
- Female
- Genotype
- Humans
- Inactivation, Metabolic/genetics
- Middle Aged
- Pharmacogenomic Variants/genetics
- Phenotype
- Polymorphism, Single Nucleotide/genetics
- Tamoxifen/administration & dosage
- Tamoxifen/adverse effects
- Tamoxifen/metabolism
Collapse
Affiliation(s)
- Yeimy Viviana Ariza Márquez
- Universidad Nacional de Colombia, Instituto de Biotecnología IBUN, Departamento de Farmacia, Bogota, 111321, Colombia
| | - Ignacio Briceño
- Universidad de la Sabana, Facultad de Medicina, Bogota, 140013, Colombia
- Pontificia Universidad Javeriana, Facultad de Medicina, Instituto de Genética Humana IGH, Bogota, 110231, Colombia
| | - Fabio Aristizábal
- Universidad Nacional de Colombia, Instituto de Biotecnología IBUN, Departamento de Farmacia, Bogota, 111321, Colombia
| | - Luis Fernando Niño
- Universidad Nacional de Colombia, Facultad de Ingeniería, Departamento de Ingeniería de Sistemas e Industrial, Bogota, 111321, Colombia
| | - Juvenal Yosa Reyes
- Universidad Simón Bolivar, Facultad de Ciencias Básicas y Biomédicas, Laboratorio de Simulación Molecular y Bioinformática, Barranquilla, 080002, Colombia.
| |
Collapse
|
15
|
Microsecond MD simulations of human CYP2D6 wild-type and five allelic variants reveal mechanistic insights on the function. PLoS One 2018; 13:e0202534. [PMID: 30133539 PMCID: PMC6104999 DOI: 10.1371/journal.pone.0202534] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/03/2018] [Indexed: 11/19/2022] Open
Abstract
Characterization of cytochrome P450 2D6 (CYP2D6) and the impact of the major identified allelic variants on the activity of one of the most dominating drug-metabolising enzymes is essential to increase drug safety and avoid adverse reactions. Microsecond molecular dynamics simulations have been performed to capture the dynamic signatures of this complex enzyme and five allelic variants with diverse enzymatic activity. In addition to the apo simulations, three substrates (bufuralol, veliparib and tamoxifen) and two inhibitors (prinomastat and quinidine) were included to explore their influence on the structure and dynamical features of the enzyme. Our results indicate that the altered enzyme activity can be attributed to changes in the hydrogen bonding network within the active site, and local structural differences in flexibility, position and shape of the binding pocket. In particular, the increased (CYP2D6*53) or the decreased (CYP2D6*17) activity seems to be related to a change in dynamics of mainly the BC loop due to a modified hydrogen bonding network around this region. In addition, the smallest active site volume was found for CYP2D6*4 (no activity). CYP2D6*2 (normal activity) showed no major differences in dynamic behaviour compared to the wild-type.
Collapse
|
16
|
Don CG, Smieško M. Out‐compute drug side effects: Focus on cytochrome P450 2D6 modeling. WILEY INTERDISCIPLINARY REVIEWS-COMPUTATIONAL MOLECULAR SCIENCE 2018. [DOI: 10.1002/wcms.1366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Charleen G. Don
- Department of Pharmaceutical SciencesUniversity of BaselBaselSwitzerland
| | - Martin Smieško
- Department of Pharmaceutical SciencesUniversity of BaselBaselSwitzerland
| |
Collapse
|
17
|
Keighobadi M, Emami S, Lagzian M, Fakhar M, Rafiei A, Valadan R. Molecular Modeling and Structural Stability of Wild-Type and Mutant CYP51 from Leishmania major: In Vitro and In Silico Analysis of a Laboratory Strain. Molecules 2018; 23:molecules23030696. [PMID: 29562710 PMCID: PMC6017637 DOI: 10.3390/molecules23030696] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 02/22/2018] [Accepted: 03/14/2018] [Indexed: 11/16/2022] Open
Abstract
Cutaneous leishmaniasis is a neglected tropical disease and a major public health in the most countries. Leishmania major is the most common cause of cutaneous leishmaniasis. In the Leishmania parasites, sterol 14α-demethylase (CYP51), which is involved in the biosynthesis of sterols, has been identified as an attractive target for development of new therapeutic agents. In this study, the sequence and structure of CYP51 in a laboratory strain (MRHO/IR/75/ER) of L. major were determined and compared to the wild-type strain. The results showed 19 mutations including seven non-synonymous and 12 synonymous ones in the CYP51 sequence of strain MRHO/IR/75/ER. Importantly, an arginine to lysine substitution at position of 474 resulted in destabilization of CYP51 (ΔΔG = 1.17 kcal/mol) in the laboratory strain; however, when the overall effects of all substitutions were evaluated by 100 ns molecular dynamics simulation, the final structure did not show any significant changes (p-value < 0.05) in stability parameter of the strain MRHO/IR/75/ER compared to the wild-type protein. The energy level for the CYP51 of wild-type and MRHO/IR/75/ER strain were −40,027.1 and −39,706.48 Kcal/mol respectively. The overall Root-mean-square deviation (RMSD) deviation between two proteins was less than 1 Å throughout the simulation and Root-mean-square fluctuation (RMSF) plot also showed no substantial differences between amino acids fluctuation of the both protein. The results also showed that, these mutations were located on the protein periphery that neither interferes with protein folding nor with substrate/inhibitor binding. Therefore, L. major strain MRHO/IR/75/ER is suggested as a suitable laboratory model for studying biological role of CYP51 and inhibitory effects of sterol 14α-demethylase inhibitors.
Collapse
Affiliation(s)
- Masoud Keighobadi
- Pharmaceutical Sciences Research Center, Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran.
| | - Saeed Emami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, 48157-33971, Iran.
| | - Milad Lagzian
- Department of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan 98168-76578, Iran.
| | - Mahdi Fakhar
- Department of Parasitology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran.
- Molecular and Cell Biology Research Center (MCBRC), Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran.
| | - Alireza Rafiei
- Molecular and Cell Biology Research Center (MCBRC), Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran.
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran.
| | - Reza Valadan
- Molecular and Cell Biology Research Center (MCBRC), Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran.
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran.
| |
Collapse
|
18
|
Zhan XY, Zhu QY. Evolution of methicillin-resistant Staphylococcus aureus: Evidence of positive selection in a penicillin-binding protein (PBP) 2a coding gene mecA. INFECTION GENETICS AND EVOLUTION 2018; 59:16-22. [PMID: 29413881 DOI: 10.1016/j.meegid.2018.01.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 01/19/2018] [Accepted: 01/22/2018] [Indexed: 11/15/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (S. aureus) (MRSA) represents more and more S. aureus infections. MecA, the novel coding gene of penicillin-binding protein (PBP) 2a of MRSA, is the key resistance factor of β-lactam, but little is known about the evolution of this gene. Given the crucial role of mecA in S. aureus physiology and β-lactam resistance, the selective forces may contribute to adaptation of the bacteria to the special environments such as its host or antibiotics. To understand the evolution of this gene, we screened GenBank database and analyzed mecA of 249 S. aureus strains. Twenty-nine unique alleles with 26 unique amino acid sequences were identified. Phylogenetic analysis showed three main groups of mecA in the global S. aureus strains. Analysis of these alleles using codon-substitution models (M8, M3, and M2a) and likelihood ratio tests (LRTs) of the codeML package and a random-effects likelihood (REL) method of HyPhy package for the site-specific ratio of nonsynonymous to synonymous substitution rates suggested that fourteen sites in the allosteric domain of PBP2a have been subjected to strong positive selection pressure. Mutations of two positive selection sites (N146K and E239K) were reported to be essential for ceftaroline- or L-695, 256-resistant. Further study indicated that the positive selection pressure might be more likely related to the host's inflammatory or immune response during S. aureus infection. Our studies provide the first evidence of positive Darwinian selection in the mecA of S. aureus, contributing to a better understanding of the adaptive mechanism of this bacterium.
Collapse
Affiliation(s)
- Xiao-Yong Zhan
- Guangzhou KingMed Center for Clinical Laboratory, Guangzhou 510300, China; KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 510300, China; The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Qing-Yi Zhu
- Guangzhou KingMed Center for Clinical Laboratory, Guangzhou 510300, China; KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 510300, China
| |
Collapse
|
19
|
Bank PCD, Swen JJ, Guchelaar HJ. Implementation of Pharmacogenomics in Everyday Clinical Settings. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2018; 83:219-246. [PMID: 29801576 DOI: 10.1016/bs.apha.2018.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Currently, germline pharmacogenomics (PGx) is successfully implemented within certain specialties in clinical care. With the integration of PGx in pharmacotherapy multiple stakeholders are involved, which are identified in this chapter. Clinically relevant pharmacogenes with their related PGx test are discussed, along with diagnostic test criteria to guide clinicians and policy makers in PGx test selection. The chapter further reviews the similarities and the differences between the guidelines of the Dutch Pharmacogenetics Working Group and the Clinical Pharmacogenetics Implementation Consortium which both support healthcare professionals in understanding PGx test results and help guiding pharmacotherapy by providing evidence-based dosing recommendations. Finally, clinical studies which provide scientific evidence and information on cost-effectiveness supporting clinical implementation of PGx in clinical care are discussed along with the remaining barriers for adoption of PGx testing by healthcare professionals.
Collapse
Affiliation(s)
- Paul C D Bank
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
20
|
LIU LIJUN, CHANG YU, DU SHULI, SHI XUGANG, YANG HUA, KANG LONGLI, JIN TIANBO, YUAN DONGYA, HE YONGJUN. Genetic analysis of drug metabolizing phase-I enzymes CYP3A4 in Tibetan populations. J Genet 2017; 96:219-225. [DOI: 10.1007/s12041-017-0757-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
21
|
Watanabe Y, Fukuyoshi S, Kato K, Hiratsuka M, Yamaotsu N, Hirono S, Gouda H, Oda A. Investigation of substrate recognition for cytochrome P450 1A2 mediated by water molecules using docking and molecular dynamics simulations. J Mol Graph Model 2017; 74:326-336. [PMID: 28475969 DOI: 10.1016/j.jmgm.2017.04.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/07/2017] [Accepted: 04/06/2017] [Indexed: 02/08/2023]
Abstract
The role of water molecules in the active site of cytochrome P450 1A2 (CYP1A2) was investigated using an explicit water model to simulate biological environments. Moreover, differences in ligand recognition between the inhibitor α-naphthoflavone (ANF) and the substrate 7-ethoxyresorufin (7ER) in the CYP1A2 complex were examined. More than 200-ns molecular dynamics (MD) simulations were performed for each complex structure of CYP1A2. In the complex structure with 7ER obtained after MD simulation, some water molecules existed in the active site and formed hydrogen bonds between 7ER and some residues. However, in the complex structure with ANF, the hydrogen bond network differed. These results suggest that CYP1A2 requires water molecules in its active site for substrate recognition. The observed differences in the hydrogen bond network in the complex with ANF or 7ER may be due to the fact that ANF is an inhibitor.
Collapse
Affiliation(s)
- Yurie Watanabe
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan; School of Pharmacy, Showa University, Shinagawa-ku, Tokyo, Japan
| | - Shuichi Fukuyoshi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Koichi Kato
- Graduate School of Pharmacy, Meijo University, Tempaku-ku, Nagoya, Aichi, Japan
| | - Masahiro Hiratsuka
- Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | | | - Shuichi Hirono
- School of Pharmacy, Kitasato University, Minato-ku, Tokyo, Japan
| | - Hiroaki Gouda
- School of Pharmacy, Showa University, Shinagawa-ku, Tokyo, Japan
| | - Akifumi Oda
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan; Graduate School of Pharmacy, Meijo University, Tempaku-ku, Nagoya, Aichi, Japan; Institute for Protein Research, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
22
|
Issa NT, Wathieu H, Ojo A, Byers SW, Dakshanamurthy S. Drug Metabolism in Preclinical Drug Development: A Survey of the Discovery Process, Toxicology, and Computational Tools. Curr Drug Metab 2017; 18:556-565. [PMID: 28302026 PMCID: PMC5892202 DOI: 10.2174/1389200218666170316093301] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/16/2016] [Accepted: 01/17/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND While establishing efficacy in translational models and humans through clinically-relevant endpoints for disease is of great interest, assessing the potential toxicity of a putative therapeutic drug is critical. Toxicological assessments in the pre-clinical discovery phase help to avoid future failure in the clinical phases of drug development. Many in vitro assays exist to aid in modular toxicological assessment, such as hepatotoxicity and genotoxicity. While these methods have provided tremendous insight into human toxicity by investigational new drugs, they are expensive, require substantial resources, and do not account for pharmacogenomics as well as critical ADME properties. Computational tools can fill this niche in toxicology if in silico models are accurate in relating drug molecular properties to toxicological endpoints as well as reliable in predicting important drug-target interactions that mediate known adverse events or adverse outcome pathways (AOPs). METHODS We undertook an unstructured search of multiple bibliographic databases for peer-reviewed literature regarding computational methods in predictive toxicology for in silico drug discovery. As this review paper is meant to serve as a survey of available methods for the interested reader, no focused criteria were applied. Literature chosen was based on the writers' expertise and intent in communicating important aspects of in silico toxicology to the interested reader. CONCLUSION This review provides a purview of computational methods of pre-clinical toxicologic assessments for novel small molecule drugs that may be of use for novice and experienced investigators as well as academic and commercial drug discovery entities.
Collapse
Affiliation(s)
- Naiem T. Issa
- Georgetown-Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University Medical Center, Washington DC, 20057 USA
| | - Henri Wathieu
- Georgetown-Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University Medical Center, Washington DC, 20057 USA
| | - Abiola Ojo
- College of Pharmacy, Howard University, Washington, DC 20059, USA
| | - Stephen W. Byers
- Georgetown-Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University Medical Center, Washington DC, 20057 USA
- Department of Biochemistry & Molecular Biology, Georgetown University, Washington DC, 20057, USA
| | - Sivanesan Dakshanamurthy
- Georgetown-Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University Medical Center, Washington DC, 20057 USA
- Department of Biochemistry & Molecular Biology, Georgetown University, Washington DC, 20057, USA
| |
Collapse
|