1
|
Huang Y, Tan H, Li Q, Wu X, Guo Z, Chen J, Zhang L, Diao Y. Molecular cloning and functional characterization of the shikimate kinase gene from Baphicacanthus cusia. FRONTIERS IN PLANT SCIENCE 2025; 16:1560891. [PMID: 40353225 PMCID: PMC12062003 DOI: 10.3389/fpls.2025.1560891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/17/2025] [Indexed: 05/14/2025]
Abstract
Baphicacanthus cusia (Nee) Bremek, a perennial herbaceous plant with medicinal properties, has limited genomic insights regarding the genes involved in its indole alkaloid biosynthesis pathway. In this study, the BcSK gene was isolated and cloned from the transcriptome data of B. cusia. The full-length cDNA of BcSK is 1,657 bp, comprising a 265 bp 5' UTR, a 507 bp 3' UTR, and an 885 bp ORF encoding 295 amino acids. The exon-intron structure of BcSK consists of four exons and three introns. Bioinformatics and phylogenetic analyses revealed a high degree of homology between BcSK and its counterparts in various plant species. Quantitative real-time polymerase chain reaction (RT-qPCR) analysis showed that BcSK expression was significantly altered under abiotic stress conditions, including methyl jasmonate (MeJA), abscisic acid (ABA), and ultraviolet (UV) radiation. The gene was predominantly expressed in flowers compared to roots, stems, and leaves. Subcellular localization analysis indicated that BcSK is primarily expressed in chloroplasts, confirming that the conversion of shikimic acid to shikimate-3-phosphate occurs in this organelle. Prokaryotic expression and enzyme activity assays demonstrated that the heterologously expressed BcSK protein catalyzed the conversion of shikimic acid to shikimate-3-phosphate. Furthermore, the ectopic overexpression of BcSK in Isatis indigotica significantly enhanced the biosynthetic flux toward indole alkaloids, including indole, indigo, and indirubin. In conclusion, this study identifies and characterizes a novel BcSK gene, providing new insights and potential applications for the metabolic engineering of B. cusia.
Collapse
Affiliation(s)
- Yuxiang Huang
- School of Pharmacy, Quanzhou Medical College, Quanzhou, China
- School of Medicine, Huaqiao University, Quanzhou, China
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Hexin Tan
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Qing Li
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Xunxun Wu
- School of Medicine, Huaqiao University, Quanzhou, China
| | - Zhiying Guo
- School of Medicine, Huaqiao University, Quanzhou, China
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Junfeng Chen
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Zhang
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Yong Diao
- School of Medicine, Huaqiao University, Quanzhou, China
| |
Collapse
|
2
|
Freitas de Freitas T, Roth CD, Abbadi BL, Hopf FSM, Perelló MA, de Matos Czeczot A, de Souza EV, Borsoi AF, Machado P, Bizarro CV, Basso LA, Timmers LFSM. Identification of potential inhibitors of Mycobacterium tuberculosis shikimate kinase: molecular docking, in silico toxicity and in vitro experiments. J Comput Aided Mol Des 2023; 37:117-128. [PMID: 36547753 PMCID: PMC9772590 DOI: 10.1007/s10822-022-00495-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Tuberculosis (TB) is one of the main causes of death from a single pathological agent, Mycobacterium tuberculosis (Mtb). In addition, the emergence of drug-resistant TB strains has exacerbated even further the treatment outcome of TB patients. It is thus needed the search for new therapeutic strategies to improve the current treatment and to circumvent the resistance mechanisms of Mtb. The shikimate kinase (SK) is the fifth enzyme of the shikimate pathway, which is essential for the survival of Mtb. The shikimate pathway is absent in humans, thereby indicating SK as an attractive target for the development of anti-TB drugs. In this work, a combination of in silico and in vitro techniques was used to identify potential inhibitors for SK from Mtb (MtSK). All compounds of our in-house database (Centro de Pesquisas em Biologia Molecular e Funcional, CPBMF) were submitted to in silico toxicity analysis to evaluate the risk of hepatotoxicity. Docking experiments were performed to identify the potential inhibitors of MtSK according to the predicted binding energy. In vitro inhibitory activity of MtSK-catalyzed chemical reaction at a single compound concentration was assessed. Minimum inhibitory concentration values for in vitro growth of pan-sensitive Mtb H37Rv strain were also determined. The mixed approach implemented in this work was able to identify five compounds that inhibit both MtSK and the in vitro growth of Mtb.
Collapse
Affiliation(s)
- Talita Freitas de Freitas
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF), Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90616-900, Brazil
| | - Candida Deves Roth
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF), Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Bruno Lopes Abbadi
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF), Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Fernanda Souza Macchi Hopf
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF), Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90616-900, Brazil
| | - Marcia Alberton Perelló
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF), Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Alexia de Matos Czeczot
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF), Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90616-900, Brazil
| | - Eduardo Vieira de Souza
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF), Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90616-900, Brazil
| | - Ana Flávia Borsoi
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF), Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90616-900, Brazil
| | - Pablo Machado
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF), Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90616-900, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90616-900, Brazil
| | - Cristiano Valim Bizarro
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF), Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90616-900, Brazil
| | - Luiz Augusto Basso
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF), Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90616-900, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90616-900, Brazil
| | - Luis Fernando Saraiva Macedo Timmers
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF), Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil. .,Universidade do Vale do Taquari (Univates), Lajeado, Rio Grande do Sul, 95914-014, Brazil. .,Programa de Pós-Graduação em Biotecnologia, Universidade do Vale do Taquari (Univates), Lajeado, Rio Grande do Sul, 95914-014, Brazil. .,Programa de Pós-Graduação em Ciências Médicas, Universidade do Vale do Taquari (Univates), Lajeado, Rio Grande do Sul, 95914-014, Brazil.
| |
Collapse
|
3
|
Rajput VS, Runthala A, Khan IA. Shikimate Kinase Inhibitors: An Update on Promising Strategy against Mycobacterium tuberculosis. Curr Drug Targets 2023; 24:388-405. [PMID: 36752299 DOI: 10.2174/1389450124666230208102645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 11/16/2022] [Accepted: 11/25/2022] [Indexed: 02/09/2023]
Abstract
Humanity has been battling with tuberculosis (TB) for a long period, and despite the availability of drugs well-known to act against the deadly microbe, the menace is still very far from reaching its end. Moreover, problems related to TB chemotherapy, such as lengthy treatment periods leading to poor patient compliance, increasing drug resistance, and association with another deadlier disease HIV-AIDS, make the situation alarming, thereby pressing the need for the discovery of new potent drugs urgently. Therefore, a drug target that is essential for survival and exclusive to M. tuberculosis presents a promising platform to explore novel molecules against the microorganism for better pathogen clearance with minimal toxicity. The shikimate pathway that leads to the synthesis of essential aromatic amino acids is one such attractive target. Shikimate kinase, the fifth enzyme of this pathway, converts shikimate to shikimate-3-phosphate by using ATP as a cosubstrate. Targeting shikimate kinase could be an effective strategy in light of its essentiality and absence of any homologue in mammals. This review discusses different strategies adopted for discovering novel compounds or scaffolds targeting M. tuberculosis shikimate kinase (MtSK) in vitro. The application of substrate analogues, their structure, and ligand-based approach for screening a library of anti-mycobacterial compounds, marine-derived molecules, and commercially available libraries have yielded promising MtSK inhibitors exhibiting micro-molar activities. To develop these leads into future drugs with minimum off-target effects on the host microenvironment, the molecules need to be structurally optimized for improved activities against enzymes and whole-cell organisms.
Collapse
Affiliation(s)
- Vikrant Singh Rajput
- Department of Biomedical Engineering, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, 305817, Ajmer, Rajasthan, India
| | - Ashish Runthala
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India
| | - Inshad Ali Khan
- Department of Microbiology, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, 305817, Ajmer, Rajasthan, India
| |
Collapse
|
4
|
Rahul Reddy MB, Krishnasamy SK, Kathiravan MK. Identification of novel scaffold using ligand and structure based approach targeting shikimate kinase. Bioorg Chem 2020; 102:104083. [PMID: 32745735 DOI: 10.1016/j.bioorg.2020.104083] [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] [Received: 04/01/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 10/23/2022]
Abstract
Tuberculosis (TB) remains a major global health problem. It causes ill-health among millions of people each year and rank as the second leading cause of death from an infectious disease worldwide, after the human immunodeficiency virus (HIV). Shikimate kinase is one of the major enzymes targeted for TB. Most approaches to overcome TB were based on synthesis and screening of a known compounds to obtain a few representatives with desired potency. In this study, we have applied a virtual screening approach which combines ligand- and structure-based approaches to screen a large library of compounds as a starting point for the identification of new scaffolds for the development of shikimate kinase inhibitors. The combined approach has identified 2 new scaffolds as potential inhibitors of shikimate kinase. To prove the approach, few of the molecules and their derivatives, a total of 17 compounds, were synthesized. The compounds were tested for biological activity and shows moderate activity against shikimate kinase. The shikimate kinase enzyme inhibition study reveals that the compounds showed inhibition (IC50) at concentrations of 50 µg/mL (Compounds 21, 22, 24, 25, 26, 27, 30, 32, 34) and 25 µg/mL (14, 19, 23, 31, 33).
Collapse
Affiliation(s)
- M B Rahul Reddy
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM IST Kattankulathur, Kancheepuram, Tamil Nadu 603203, India
| | | | - M K Kathiravan
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM IST Kattankulathur, Kancheepuram, Tamil Nadu 603203, India; 209, Dr APJ Abdul Kalam Research Lab, SRM College of Pharmacy, SRM IST Kattankulathur, Kancheepuram, Tamil Nadu 603203, India
| |
Collapse
|
5
|
Mycobacterium tuberculosis Shikimate Pathway Enzymes as Targets for the Rational Design of Anti-Tuberculosis Drugs. Molecules 2020; 25:molecules25061259. [PMID: 32168746 PMCID: PMC7144000 DOI: 10.3390/molecules25061259] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 12/17/2022] Open
Abstract
Roughly a third of the world’s population is estimated to have latent Mycobacterium tuberculosis infection, being at risk of developing active tuberculosis (TB) during their lifetime. Given the inefficacy of prophylactic measures and the increase of drug-resistant M. tuberculosis strains, there is a clear and urgent need for the development of new and more efficient chemotherapeutic agents, with selective toxicity, to be implemented on patient treatment. The component enzymes of the shikimate pathway, which is essential in mycobacteria and absent in humans, stand as attractive and potential targets for the development of new drugs to treat TB. This review gives an update on published work on the enzymes of the shikimate pathway and some insight on what can be potentially explored towards selective drug development.
Collapse
|
6
|
Favela-Candia A, Téllez-Valencia A, Campos-Almazán M, Sierra-Campos E, Valdez-Solana M, Oria-Hernández J, Castillo-Villanueva A, Nájera H, Avitia-Domínguez C. Biochemical, Kinetic, and Computational Structural Characterization of Shikimate Kinase from Methicillin-Resistant Staphylococcus aureus. Mol Biotechnol 2019; 61:274-285. [PMID: 30747382 DOI: 10.1007/s12033-019-00159-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
One of the most widespread pathogens worldwide is methicillin-resistant Staphylococcus aureus, a bacterium that provokes severe life-threatening illnesses both in hospitals and in the community. The principal challenge lies in the resistance of MRSA to current treatments, which encourages the study of different molecular targets that could be used to develop new drugs against this infectious agent. With this goal, a detailed characterization of shikimate kinase from this microorganism (SaSK) is described. The results showed that SaSK has a Km of 0.153 and 224 µM for shikimate and ATP, respectively, and a global reaction rate of 13.4 µmol/min/mg; it is suggested that SaSK utilizes the Bi-Bi Ping Pong reaction mechanism. Furthermore, the physicochemical data indicated that SaSK is an unstable, hydrophilic, and acidic protein. Finally, structural information showed that SaSK presented folding that is typical of its homologous counterparts and contains the typical domains of this family of proteins. Amino acids that have been shown to be important for SaSK protein function are conserved. Therefore, this study provides fundamental information that may aid in the design of inhibitors that could be used to develop new antibacterial agents.
Collapse
Affiliation(s)
- Alejandro Favela-Candia
- Facultad de Medicina y Nutrición, Universidad Juárez del Estado de Durango, Av. Universidad y Fanny Anitua S/N, C.P. 34000, Durango, Dgo, Mexico
| | - Alfredo Téllez-Valencia
- Facultad de Medicina y Nutrición, Universidad Juárez del Estado de Durango, Av. Universidad y Fanny Anitua S/N, C.P. 34000, Durango, Dgo, Mexico
| | - Mara Campos-Almazán
- Facultad de Medicina y Nutrición, Universidad Juárez del Estado de Durango, Av. Universidad y Fanny Anitua S/N, C.P. 34000, Durango, Dgo, Mexico
| | - Erick Sierra-Campos
- Facultad de Ciencias Químicas, Universidad Juárez del Estado de Durango, Av. Artículo 123 S/N Fracc. Filadelfia, Gómez Palacio, C.P. 35010, Durango, Mexico
| | - Mónica Valdez-Solana
- Facultad de Ciencias Químicas, Universidad Juárez del Estado de Durango, Av. Artículo 123 S/N Fracc. Filadelfia, Gómez Palacio, C.P. 35010, Durango, Mexico
| | - Jesús Oria-Hernández
- Laboratorio de Bioquímica Genética, Secretaría de Salud, Instituto Nacional de Pediatría, C.P. 04534, Ciudad de México, Mexico
| | - Adriana Castillo-Villanueva
- Laboratorio de Bioquímica Genética, Secretaría de Salud, Instituto Nacional de Pediatría, C.P. 04534, Ciudad de México, Mexico
| | - Hugo Nájera
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, Delegación Cuajimalpa de Morelos, Av. Vasco de Quiroga 4871, Colonia Santa Fe Cuajimalpa, C.P. 05300, Ciudad de México, Mexico
| | - Claudia Avitia-Domínguez
- Facultad de Medicina y Nutrición, Universidad Juárez del Estado de Durango, Av. Universidad y Fanny Anitua S/N, C.P. 34000, Durango, Dgo, Mexico.
| |
Collapse
|
7
|
Simithy J, Fuanta NR, Alturki M, Hobrath JV, Wahba AE, Pina I, Rath J, Hamann MT, DeRuiter J, Goodwin DC, Calderón AI. Slow-Binding Inhibition of Mycobacterium tuberculosis Shikimate Kinase by Manzamine Alkaloids. Biochemistry 2018; 57:4923-4933. [PMID: 30063132 DOI: 10.1021/acs.biochem.8b00231] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tuberculosis represents a significant public health crisis. There is an urgent need for novel molecular scaffolds against this pathogen. We screened a small library of marine-derived compounds against shikimate kinase from Mycobacterium tuberculosis ( MtSK), a promising target for antitubercular drug development. Six manzamines previously shown to be active against M. tuberculosis were characterized as MtSK inhibitors: manzamine A (1), 8-hydroxymanzamine A (2), manzamine E (3), manzamine F (4), 6-deoxymanzamine X (5), and 6-cyclohexamidomanzamine A (6). All six showed mixed noncompetitive inhibition of MtSK. The lowest KI values were obtained for 6 across all MtSK-substrate complexes. Time-dependent analyses revealed two-step, slow-binding inhibition. The behavior of 1 was typical; initial formation of an enzyme-inhibitor complex (EI) obeyed an apparent KI of ∼30 μM with forward ( k5) and reverse ( k6) rate constants for isomerization to an EI* complex of 0.18 and 0.08 min-1, respectively. In contrast, 6 showed a lower KI for the initial encounter complex (∼1.5 μM), substantially faster isomerization to EI* ( k5 = 0.91 min-1), and slower back conversion of EI* to EI ( k6 = 0.04 min-1). Thus, the overall inhibition constants, KI*, for 1 and 6 were 10 and 0.06 μM, respectively. These findings were consistent with docking predictions of a favorable binding mode and a second, less tightly bound pose for 6 at MtSK. Our results suggest that manzamines, in particular 6, constitute a new scaffold from which drug candidates with novel mechanisms of action could be designed for the treatment of tuberculosis by targeting MtSK.
Collapse
Affiliation(s)
- Johayra Simithy
- Department of Drug Discovery and Development, Harrison School of Pharmacy , Auburn University , 4306 Walker Building , Auburn , Alabama 36849 , United States
| | - Ngolui Rene Fuanta
- Department of Chemistry and Biochemistry , Auburn University , 179 Chemistry Building , Auburn , Alabama 36849 , United States
| | - Mansour Alturki
- Department of Drug Discovery and Development, Harrison School of Pharmacy , Auburn University , 4306 Walker Building , Auburn , Alabama 36849 , United States
| | - Judith V Hobrath
- Department of Chemistry , University of Alabama at Birmingham , Birmingham , Alabama 35294 , United States
| | - Amir E Wahba
- Chemistry Department, Faculty of Science , Damietta University , Damietta , Egypt
| | - Ivett Pina
- Departments of Drug Discovery & Biomedical Sciences and Public Health, Colleges of Pharmacy and Medicine , The Medical University of South Carolina , 70 President Street, MSP 139 , Charleston , South Carolina 29425 , United States
| | - Jnanendra Rath
- Department of Botany , Visva-Bharati University , Santiniketan , West Bengal 731235 , India
| | - Mark T Hamann
- Departments of Drug Discovery & Biomedical Sciences and Public Health, Colleges of Pharmacy and Medicine , The Medical University of South Carolina , 70 President Street, MSP 139 , Charleston , South Carolina 29425 , United States
| | - Jack DeRuiter
- Department of Drug Discovery and Development, Harrison School of Pharmacy , Auburn University , 4306 Walker Building , Auburn , Alabama 36849 , United States
| | - Douglas C Goodwin
- Department of Chemistry and Biochemistry , Auburn University , 179 Chemistry Building , Auburn , Alabama 36849 , United States
| | - Angela I Calderón
- Department of Drug Discovery and Development, Harrison School of Pharmacy , Auburn University , 4306 Walker Building , Auburn , Alabama 36849 , United States
| |
Collapse
|
8
|
Schoenenberger B, Wszolek A, Meier R, Brundiek H, Obkircher M, Wohlgemuth R. Recombinant AroL-Catalyzed Phosphorylation for the Efficient Synthesis of Shikimic Acid 3-Phosphate. Biotechnol J 2018; 13:e1700529. [PMID: 29697210 DOI: 10.1002/biot.201700529] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 04/03/2018] [Indexed: 01/01/2023]
Abstract
Shikimic acid 3-phosphate, as a central metabolite of the shikimate pathway, is of high interest as enzyme substrate for 5-enolpyruvoyl-shikimate 3-phosphate synthase, a drug target in infectious diseases and a prime enzyme target for the herbicide glyphosate. As the important substrate shikimic acid 3-phosphate is only accessible via a chemical multi-step route, a new straightforward preparative one-step enzymatic phosphorylation of shikimate using a stable recombinant shikimate kinase has been developed for the selective phosphorylation of shikimate in the 3-position. Highly active shikimate kinase is produced by straightforward expression of a synthetic aroL gene in Escherichia coli. The time course of the shikimate kinase-catalyzed phosphorylation is investigated by 1 H- and 31 P-NMR, using the phosphoenolpyruvate/pyruvate kinase system for the regeneration of the ATP cofactor. This enables the development of a quantitative biocatalytic 3-phosphorylation of shikimic acid. After a standard workup procedure, a good yield of shikimic acid 3-phosphate, with high HPLC- and NMR purity, is obtained. This efficient biocatalytic synthesis of shikimic acid 3-phosphate is superior to any other method and has been successfully scaled up to multi-gram scale.
Collapse
Affiliation(s)
| | - Agata Wszolek
- Enzymicals, Walther-Rathenau-Strasse 49a, 17489, Greifswald, Germany
| | - Roland Meier
- Sigma-Aldrich, Member of Merck Group, Industriestrasse 25, CH-9470, Buchs, Switzerland
| | - Henrike Brundiek
- Enzymicals, Walther-Rathenau-Strasse 49a, 17489, Greifswald, Germany
| | - Markus Obkircher
- Sigma-Aldrich, Member of Merck Group, Industriestrasse 25, CH-9470, Buchs, Switzerland
| | - Roland Wohlgemuth
- Sigma-Aldrich, Member of Merck Group, Industriestrasse 25, CH-9470, Buchs, Switzerland
| |
Collapse
|
9
|
Yao J, Wang X, Luo H, Gu P. Understanding the Catalytic Mechanism and the Nature of the Transition State of an Attractive Drug‐Target Enzyme (Shikimate Kinase) by Quantum Mechanical/Molecular Mechanical (QM/MM) Studies. Chemistry 2017; 23:16380-16387. [DOI: 10.1002/chem.201703867] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Indexed: 11/05/2022]
Affiliation(s)
- Jianzhuang Yao
- School of Biological Science and Technology University of Jinan Jinan 250022 P.R. China
| | - Xia Wang
- School of Biological Science and Technology University of Jinan Jinan 250022 P.R. China
| | - Haixia Luo
- Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Life Science School Ningxia University Yinchuan 750021 P.R. China
| | - Pengfei Gu
- School of Biological Science and Technology University of Jinan Jinan 250022 P.R. China
| |
Collapse
|
10
|
Protein kinase C-δ inhibitor, Rottlerin inhibits growth and survival of mycobacteria exclusively through Shikimate kinase. Biochem Biophys Res Commun 2016; 478:721-6. [DOI: 10.1016/j.bbrc.2016.08.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 08/03/2016] [Indexed: 02/07/2023]
|
11
|
Mehra R, Rajput VS, Gupta M, Chib R, Kumar A, Wazir P, Khan IA, Nargotra A. Benzothiazole Derivative as a Novel Mycobacterium tuberculosis Shikimate Kinase Inhibitor: Identification and Elucidation of Its Allosteric Mode of Inhibition. J Chem Inf Model 2016; 56:930-40. [PMID: 27149193 DOI: 10.1021/acs.jcim.6b00056] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Mycobacterium tuberculosis shikimate kinase (Mtb-SK) is a key enzyme involved in the biosynthesis of aromatic amino acids through the shikimate pathway. Since it is proven to be essential for the survival of the microbe and is absent from mammals, it is a promising target for anti-TB drug discovery. In this study, a combined approach of in silico similarity search and pharmacophore building using already reported inhibitors was used to screen a procured library of 20,000 compounds of the commercially available ChemBridge database. From the in silico screening, 15 hits were identified, and these hits were evaluated in vitro for Mtb-SK enzyme inhibition. Two compounds presented significant enzyme inhibition with IC50 values of 10.69 ± 0.9 and 46.22 ± 1.2 μM. The best hit was then evaluated for the in vitro mode of inhibition where it came out to be an uncompetitive and noncompetitive inhibitor with respect to shikimate (SKM) and ATP, respectively, suggesting its binding at an allosteric site. Potential binding sites of Mtb-SK were identified which confirmed the presence of an allosteric binding pocket apart from the ATP and SKM binding sites. The docking simulations were performed at this pocket in order to find the mode of binding of the best hit in the presence of substrates and the products of the enzymatic reaction. Molecular dynamics (MD) simulations elucidated the probability of inhibitor binding at the allosteric site in the presence of ADP and shikimate-3-phosphate (S-3-P), that is, after the formation of products of the reaction. The inhibitor binding may prevent the release of the product from Mtb-SK, thereby inhibiting its activity. The binding stability and the key residue interactions of the inhibitor to this product complex were also revealed by the MD simulations. Residues ARG43, ILE45, and PHE57 were identified as crucial that were involved in interactions with the best hit. This is the first report of an allosteric binding site of Mtb-SK, which could largely address the selectivity issue associated with kinase inhibitors.
Collapse
Affiliation(s)
- Rukmankesh Mehra
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu 180001, India
| | - Vikrant Singh Rajput
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu 180001, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu 180001, India
| | - Monika Gupta
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu 180001, India
| | - Reena Chib
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu 180001, India
| | - Amit Kumar
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu 180001, India
| | - Priya Wazir
- Instrumentation Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu 180001, India
| | - Inshad Ali Khan
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu 180001, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu 180001, India
| | - Amit Nargotra
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu 180001, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu 180001, India
| |
Collapse
|
12
|
Kaur P, Datta S, Shandil RK, Kumar N, Robert N, Sokhi UK, Guptha S, Narayanan S, Anbarasu A, Ramaiah S. Unravelling the Secrets of Mycobacterial Cidality through the Lens of Antisense. PLoS One 2016; 11:e0154513. [PMID: 27144597 PMCID: PMC4856384 DOI: 10.1371/journal.pone.0154513] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/14/2016] [Indexed: 01/13/2023] Open
Abstract
One of the major impediments in anti-tubercular drug discovery is the lack of a robust grammar that governs the in-vitro to the in-vivo translation of efficacy. Mycobacterium tuberculosis (Mtb) is capable of growing both extracellular as well as intracellular; encountering various hostile conditions like acidic milieu, free radicals, starvation, oxygen deprivation, and immune effector mechanisms. Unique survival strategies of Mtb have prompted researchers to develop in-vitro equivalents to simulate in-vivo physiologies and exploited to find efficacious inhibitors against various phenotypes. Conventionally, the inhibitors are screened on Mtb under the conditions that are unrelated to the in-vivo disease environments. The present study was aimed to (1). Investigate cidality of Mtb targets using a non-chemical inhibitor antisense-RNA (AS-RNA) under in-vivo simulated in-vitro conditions.(2). Confirm the cidality of the targets under in-vivo in experimental tuberculosis. (3). Correlate in-vitro vs. in-vivo cidality data to identify the in-vitro condition that best predicts in-vivo cidality potential of the targets. Using cidality as a metric for efficacy, and AS-RNA as a target-specific inhibitor, we delineated the cidality potential of five target genes under six different physiological conditions (replicating, hypoxia, low pH, nutrient starvation, nitrogen depletion, and nitric oxide).In-vitro cidality confirmed in experimental tuberculosis in BALB/c mice using the AS-RNA allowed us to identify cidal targets in the rank order of rpoB>aroK>ppk>rpoC>ilvB. RpoB was used as the cidality control. In-vitro and in-vivo studies feature aroK (encoding shikimate kinase) as an in-vivo mycobactericidal target suitable for anti-TB drug discovery. In-vitro to in-vivo cidality correlations suggested the low pH (R = 0.9856) in-vitro model as best predictor of in-vivo cidality; however, similar correlation studies in pathologically relevant (Kramnik) mice are warranted. In the acute infection phase for the high fidelity translation, the compound efficacy may also be evaluated in the low pH, in addition to the standard replication condition.
Collapse
Affiliation(s)
- Parvinder Kaur
- Research Area, Drug Discovery, AstraZeneca India Private Limited, Bangalore, India
- * E-mail:
| | | | | | - Naveen Kumar
- Research Area, Drug Discovery, AstraZeneca India Private Limited, Bangalore, India
| | - Nanduri Robert
- Research Area, Drug Discovery, AstraZeneca India Private Limited, Bangalore, India
| | - Upneet K. Sokhi
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, United States of America
| | - Supreeth Guptha
- Research Area, Drug Discovery, AstraZeneca India Private Limited, Bangalore, India
| | - Shridhar Narayanan
- Research Area, Drug Discovery, AstraZeneca India Private Limited, Bangalore, India
| | - Anand Anbarasu
- School of Biosciences and Technology, VIT University, Vellore, India
| | - Sudha Ramaiah
- School of Biosciences and Technology, VIT University, Vellore, India
| |
Collapse
|
13
|
Screening of antitubercular compound library identifies novel shikimate kinase inhibitors of Mycobacterium tuberculosis. Appl Microbiol Biotechnol 2016; 100:5415-26. [DOI: 10.1007/s00253-015-7268-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 11/26/2015] [Accepted: 12/22/2015] [Indexed: 10/22/2022]
|
14
|
Sutton KA, Breen J, MacDonald U, Beanan JM, Olson R, Russo TA, Schultz LW, Umland TC. Structure of shikimate kinase, an in vivo essential metabolic enzyme in the nosocomial pathogen Acinetobacter baumannii, in complex with shikimate. ACTA ACUST UNITED AC 2015; 71:1736-44. [PMID: 26249354 DOI: 10.1107/s139900471501189x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/22/2015] [Indexed: 11/10/2022]
Abstract
Acinetobacter baumannii is an opportunistic Gram-negative pathogen that is an important cause of healthcare-associated infections exhibiting high mortality rates. Clinical isolates of multidrug-resistant (MDR) and extremely drug-resistant (XDR) A. baumannii strains are increasingly being observed. Compounding this concern is the dearth of new antibacterial agents in late-stage development that are effective against MDR and XDR A. baumannii. As part of an effort to address these concerns, two genes (aroA and aroC) of the shikimate pathway have previously been determined to be essential for the growth and survival of A. baumannii during host infection (i.e. to be essential in vivo). This study expands upon these results by demonstrating that the A. baumannii aroK gene, encoding shikimate kinase (SK), is also essential in vivo in a rat soft-tissue infection model. The crystal structure of A. baumannii SK in complex with the substrate shikimate and a sulfate ion that mimics the binding interactions expected for the β-phosphate of ATP was then determined to 1.91 Å resolution and the enzyme kinetics were characterized. The flexible shikimate-binding domain and LID region are compared with the analogous regions in other SK crystal structures. The impact of structural differences and sequence divergence between SKs from pathogenic bacteria that may influence antibiotic-development efforts is discussed.
Collapse
Affiliation(s)
- Kristin A Sutton
- Hauptman-Woodward Medical Research Institute, Buffalo, NY 14203, USA
| | - Jennifer Breen
- Hauptman-Woodward Medical Research Institute, Buffalo, NY 14203, USA
| | - Ulrike MacDonald
- Department of Medicine and The Witebsky Center for Microbial Pathogenesis, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - Janet M Beanan
- Department of Medicine and The Witebsky Center for Microbial Pathogenesis, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - Ruth Olson
- Department of Medicine and The Witebsky Center for Microbial Pathogenesis, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - Thomas A Russo
- Department of Medicine and The Witebsky Center for Microbial Pathogenesis, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - L Wayne Schultz
- Hauptman-Woodward Medical Research Institute, Buffalo, NY 14203, USA
| | - Timothy C Umland
- Hauptman-Woodward Medical Research Institute, Buffalo, NY 14203, USA
| |
Collapse
|
15
|
Simithy J, Gill G, Wang Y, Goodwin DC, Calderón AI. Development of an ESI-LC-MS-Based Assay for Kinetic Evaluation of Mycobacterium tuberculosis Shikimate Kinase Activity and Inhibition. Anal Chem 2015; 87:2129-36. [DOI: 10.1021/ac503210n] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Johayra Simithy
- Department
of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, 4306 Walker Building, Auburn, Alabama 36849, United States
| | - Gobind Gill
- Department
of Chemistry and Biochemistry, Auburn University, 179 Chemistry Building, Auburn, Alabama 36849, United States
| | - Yu Wang
- Department
of Chemistry and Biochemistry, Auburn University, 179 Chemistry Building, Auburn, Alabama 36849, United States
| | - Douglas C. Goodwin
- Department
of Chemistry and Biochemistry, Auburn University, 179 Chemistry Building, Auburn, Alabama 36849, United States
| | - Angela I. Calderón
- Department
of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, 4306 Walker Building, Auburn, Alabama 36849, United States
| |
Collapse
|