1
|
Roster CP, LaVigne D, Milanes JE, Knight E, Anderson HD, Pizarro S, Harding EM, Morris MT, Yan VC, Pham CD, Muller F, Kwain S, Rees KC, Dominy B, Whitehead DC, Uddin MN, Millward SW, Morris JC. Enolase Inhibitors as Early Lead Therapeutics against Trypanosoma brucei. Pathogens 2023; 12:1290. [PMID: 38003754 PMCID: PMC10675445 DOI: 10.3390/pathogens12111290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/16/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Glucose metabolism is critical for the African trypanosome, Trypanosoma brucei, serving as the lone source of ATP production for the bloodstream form (BSF) parasite in the glucose-rich environment of the host blood. Recently, phosphonate inhibitors of human enolase (ENO), the enzyme responsible for the interconversion of 2-phosphoglycerate (2-PG) to phosphoenolpyruvate (PEP) in glycolysis or PEP to 2-PG in gluconeogenesis, have been developed for the treatment of glioblastoma multiforme (GBM). Here, we have tested these agents against T. brucei ENO (TbENO) and found the compounds to be potent enzyme inhibitors and trypanocides. For example, (1-hydroxy-2-oxopyrrolidin-3-yl) phosphonic acid (deoxy-SF2312) was a potent enzyme inhibitor (IC50 value of 0.60 ± 0.23 µM), while a six-membered ring-bearing phosphonate, (1-hydroxy-2-oxopiperidin-3-yl) phosphonic acid (HEX), was less potent (IC50 value of 2.1 ± 1.1 µM). An analog with a larger seven-membered ring, (1-hydroxy-2-oxoazepan-3-yl) phosphonic acid (HEPTA), was not active. Molecular docking simulations revealed that deoxy-SF2312 and HEX had binding affinities of -6.8 and -7.5 kcal/mol, respectively, while the larger HEPTA did not bind as well, with a binding of affinity of -4.8 kcal/mol. None of these compounds were toxic to BSF parasites; however, modification of enzyme-active phosphonates through the addition of pivaloyloxymethyl (POM) groups improved activity against T. brucei, with POM-modified (1,5-dihydroxy-2-oxopyrrolidin-3-yl) phosphonic acid (POMSF) and POMHEX having EC50 values of 0.45 ± 0.10 and 0.61 ± 0.08 µM, respectively. These findings suggest that HEX is a promising lead against T. brucei and that further development of prodrug HEX analogs is warranted.
Collapse
Affiliation(s)
- Colm P. Roster
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (C.P.R.); (D.L.); (J.E.M.); (E.K.); (H.D.A.); (S.P.); (E.M.H.); (M.T.M.)
| | - Danielle LaVigne
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (C.P.R.); (D.L.); (J.E.M.); (E.K.); (H.D.A.); (S.P.); (E.M.H.); (M.T.M.)
| | - Jillian E. Milanes
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (C.P.R.); (D.L.); (J.E.M.); (E.K.); (H.D.A.); (S.P.); (E.M.H.); (M.T.M.)
| | - Emily Knight
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (C.P.R.); (D.L.); (J.E.M.); (E.K.); (H.D.A.); (S.P.); (E.M.H.); (M.T.M.)
| | - Heidi D. Anderson
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (C.P.R.); (D.L.); (J.E.M.); (E.K.); (H.D.A.); (S.P.); (E.M.H.); (M.T.M.)
| | - Sabrina Pizarro
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (C.P.R.); (D.L.); (J.E.M.); (E.K.); (H.D.A.); (S.P.); (E.M.H.); (M.T.M.)
| | - Elijah M. Harding
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (C.P.R.); (D.L.); (J.E.M.); (E.K.); (H.D.A.); (S.P.); (E.M.H.); (M.T.M.)
| | - Meredith T. Morris
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (C.P.R.); (D.L.); (J.E.M.); (E.K.); (H.D.A.); (S.P.); (E.M.H.); (M.T.M.)
| | - Victoria C. Yan
- Department of Cancer Systems Imaging, UT MD Anderson Cancer Center, Houston, TX 77030, USA; (V.C.Y.); (C.-D.P.); (M.N.U.); (S.W.M.)
| | - Cong-Dat Pham
- Department of Cancer Systems Imaging, UT MD Anderson Cancer Center, Houston, TX 77030, USA; (V.C.Y.); (C.-D.P.); (M.N.U.); (S.W.M.)
| | - Florian Muller
- Sporos Bioventures, 3000 Bissonnet, Belmont Suite 5303, Houston, TX 77005, USA;
| | - Samuel Kwain
- Eukaryotic Pathogens Innovation Center, Department of Chemistry, Clemson University, Clemson, SC 29634, USA; (S.K.); (K.C.R.); (D.C.W.)
| | - Kerrick C. Rees
- Eukaryotic Pathogens Innovation Center, Department of Chemistry, Clemson University, Clemson, SC 29634, USA; (S.K.); (K.C.R.); (D.C.W.)
| | - Brian Dominy
- Department of Chemistry, Clemson University, Clemson, SC 29634, USA;
| | - Daniel C. Whitehead
- Eukaryotic Pathogens Innovation Center, Department of Chemistry, Clemson University, Clemson, SC 29634, USA; (S.K.); (K.C.R.); (D.C.W.)
| | - Md Nasir Uddin
- Department of Cancer Systems Imaging, UT MD Anderson Cancer Center, Houston, TX 77030, USA; (V.C.Y.); (C.-D.P.); (M.N.U.); (S.W.M.)
| | - Steven W. Millward
- Department of Cancer Systems Imaging, UT MD Anderson Cancer Center, Houston, TX 77030, USA; (V.C.Y.); (C.-D.P.); (M.N.U.); (S.W.M.)
| | - James C. Morris
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (C.P.R.); (D.L.); (J.E.M.); (E.K.); (H.D.A.); (S.P.); (E.M.H.); (M.T.M.)
| |
Collapse
|
2
|
Milanes JE, Suryadi J, Monaghan NP, Harding EM, Morris CS, Rozema SD, Khalifa MM, Golden JE, Phan IQ, Zigweid R, Abendroth J, Rice CA, McCord HT, Wilson S, Fenwick MK, Morris JC. Characterization of Glucokinases from Pathogenic Free-Living Amoebae. Antimicrob Agents Chemother 2022; 66:e0237321. [PMID: 35604214 PMCID: PMC9211422 DOI: 10.1128/aac.02373-21] [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] [Received: 12/16/2021] [Accepted: 04/27/2022] [Indexed: 11/20/2022] Open
Abstract
Infection with pathogenic free-living amoebae, including Naegleria fowleri, Acanthamoeba spp., and Balamuthia mandrillaris, can lead to life-threatening illnesses, primarily because of catastrophic central nervous system involvement. Efficacious treatment options for these infections are lacking, and the mortality rate due to infection is high. Previously, we evaluated the N. fowleri glucokinase (NfGlck) as a potential target for therapeutic intervention, as glucose metabolism is critical for in vitro viability. Here, we extended these studies to the glucokinases from two other pathogenic free-living amoebae, including Acanthamoeba castellanii (AcGlck) and B. mandrillaris (BmGlck). While these enzymes are similar (49.3% identical at the amino acid level), they have distinct kinetic properties that distinguish them from each other. For ATP, AcGlck and BmGlck have apparent Km values of 472.5 and 41.0 μM, while Homo sapiens Glck (HsGlck) has a value of 310 μM. Both parasite enzymes also have a higher apparent affinity for glucose than the human counterpart, with apparent Km values of 45.9 μM (AcGlck) and 124 μM (BmGlck) compared to ~8 mM for HsGlck. Additionally, AcGlck and BmGlck differ from each other and other Glcks in their sensitivity to small molecule inhibitors, suggesting that inhibitors with pan-amoebic activity could be challenging to generate.
Collapse
Affiliation(s)
- Jillian E. Milanes
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, USA
| | - Jimmy Suryadi
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, USA
| | - Neil P. Monaghan
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, USA
| | - Elijah M. Harding
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, USA
| | - Corbin S. Morris
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, USA
| | - Soren D. Rozema
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Muhammad M. Khalifa
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Jennifer E. Golden
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Isabelle Q. Phan
- Seattle Structural Genomics Center for Infectious Disease, Center for Global Infection Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Rachael Zigweid
- Seattle Structural Genomics Center for Infectious Disease, Center for Global Infection Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Jan Abendroth
- Seattle Structural Genomics Center for Infectious Disease, Center for Global Infection Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
- UCB BioSciences, Bainbridge Island, Washington, USA
| | - Christopher A. Rice
- Pharmaceutical and Biomedical Sciences, Center for Drug Discovery, College of Pharmacy, University of Georgia, Athens, Georgia, USA
| | - Hayden T. McCord
- Pharmaceutical and Biomedical Sciences, Center for Drug Discovery, College of Pharmacy, University of Georgia, Athens, Georgia, USA
| | - Stevin Wilson
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, USA
- Genomics and Bioinformatics Facility, Clemson University, Clemson, South Carolina, USA
| | - Michael K. Fenwick
- Seattle Structural Genomics Center for Infectious Disease, Center for Global Infection Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - James C. Morris
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
3
|
Dean S. Basic Biology of Trypanosoma brucei with Reference to the Development of Chemotherapies. Curr Pharm Des 2021; 27:1650-1670. [PMID: 33463458 DOI: 10.2174/1381612827666210119105008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/01/2020] [Accepted: 12/08/2020] [Indexed: 11/22/2022]
Abstract
Trypanosoma brucei are protozoan parasites that cause the lethal human disease African sleeping sickness and the economically devastating disease of cattle, Nagana. African sleeping sickness, also known as Human African Trypanosomiasis (HAT), threatens 65 million people and animal trypanosomiasis makes large areas of farmland unusable. There is no vaccine and licensed therapies against the most severe, late-stage disease are toxic, impractical and ineffective. Trypanosomes are transmitted by tsetse flies, and HAT is therefore predominantly confined to the tsetse fly belt in sub-Saharan Africa. They are exclusively extracellular and they differentiate between at least seven developmental forms that are highly adapted to host and vector niches. In the mammalian (human) host they inhabit the blood, cerebrospinal fluid (late-stage disease), skin, and adipose fat. In the tsetse fly vector they travel from the tsetse midgut to the salivary glands via the ectoperitrophic space and proventriculus. Trypanosomes are evolutionarily divergent compared with most branches of eukaryotic life. Perhaps most famous for their extraordinary mechanisms of monoallelic gene expression and antigenic variation, they have also been investigated because much of their biology is either highly unconventional or extreme. Moreover, in addition to their importance as pathogens, many researchers have been attracted to the field because trypanosomes have some of the most advanced molecular genetic tools and database resources of any model system. The following will cover just some aspects of trypanosome biology and how its divergent biochemistry has been leveraged to develop drugs to treat African sleeping sickness. This is by no means intended to be a comprehensive survey of trypanosome features. Rather, I hope to present trypanosomes as one of the most fascinating and tractable systems to do discovery biology.
Collapse
Affiliation(s)
- Samuel Dean
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom
| |
Collapse
|
4
|
Wang H, Qin Z, Yan A. Classification models and SAR analysis on CysLT1 receptor antagonists using machine learning algorithms. Mol Divers 2021; 25:1597-1616. [PMID: 33534023 DOI: 10.1007/s11030-020-10165-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/27/2020] [Indexed: 12/21/2022]
Abstract
Cysteinyl leukotrienes 1 (CysLT1) receptor is a promising drug target for rhinitis or other allergic diseases. In our study, we built classification models to predict bioactivities of CysLT1 receptor antagonists. We built a dataset with 503 CysLT1 receptor antagonists which were divided into two groups: highly active molecules (IC50 < 1000 nM) and weakly active molecules (IC50 ≥ 1000 nM). The molecules were characterized by several descriptors including CORINA descriptors, MACCS fingerprints, Morgan fingerprint and molecular SMILES. For CORINA descriptors and two types of fingerprints, we used the random forests (RF) and deep neural networks (DNN) to build models. For molecular SMILES, we used recurrent neural networks (RNN) with the self-attention to build models. The accuracies of test sets for all models reached 85%, and the accuracy of the best model (Model 2C) was 93%. In addition, we made structure-activity relationship (SAR) analyses on CysLT1 receptor antagonists, which were based on the output from the random forest models and RNN model. It was found that highly active antagonists usually contained the common substructures such as tetrazoles, indoles and quinolines. These substructures may improve the bioactivity of the CysLT1 receptor antagonists.
Collapse
Affiliation(s)
- Hongzhao Wang
- State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering, University of Chemical Technology, Beijing, People's Republic of China
| | - Zijian Qin
- State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering, University of Chemical Technology, Beijing, People's Republic of China
| | - Aixia Yan
- State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering, University of Chemical Technology, Beijing, People's Republic of China.
| |
Collapse
|
5
|
Matta CBBD, Santos-Júnior PFDS, Gonçalves VT, Araújo MVD, Queiroz ACD, Silva JKS, Silva JFMD, Padilha RJR, Alves LC, Santos FABD, Barcellos LT, Silva-Júnior EFD, Araújo-Júnior JXD, Costa JBND, Sant’Anna CMR, Alexandre-Moreira MS. In vitro and in vivo evaluation of dialkylphosphorylhydrazones against Leishmania chagasi promastigotes and amastigotes. NEW J CHEM 2021. [DOI: 10.1039/d1nj03694g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In our study, two new dialkylphosphorylhydrazones have been designed targeting activity against L. braziliensis and L. amazonensis parasites, and their mechanism of action, as well as their leishmanicidal activity against L. chagasi, was evaluated.
Collapse
Affiliation(s)
- Carolina Barbosa Brito da Matta
- Laboratory of Pharmacology and Immunity, Institute of Biological Sciences and Health, Federal University of Alagoas, Campus A.C. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, AL, Brazil
| | | | - Vinícius Tomaz Gonçalves
- Federal Center for Technology Education Celso Suckow da Fonseca (CEFET/RJ), Itaguaí 20271-110, RJ, Brazil
| | - Morgana Vital de Araújo
- Laboratory of Pharmacology and Immunity, Institute of Biological Sciences and Health, Federal University of Alagoas, Campus A.C. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, AL, Brazil
| | - Aline Cavalcanti de Queiroz
- Laboratory of Pharmacology and Immunity, Institute of Biological Sciences and Health, Federal University of Alagoas, Campus A.C. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, AL, Brazil
| | - João Kaycke Sarmento Silva
- Laboratory of Pharmacology and Immunity, Institute of Biological Sciences and Health, Federal University of Alagoas, Campus A.C. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, AL, Brazil
| | - João Flávio Monteiro da Silva
- Laboratory of Pharmacology and Immunity, Institute of Biological Sciences and Health, Federal University of Alagoas, Campus A.C. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, AL, Brazil
| | - Rafael José Ribeiro Padilha
- Laboratory of Immunopathology Keizo Asami (LIKA) and Biochemistry Department, Federal University of Pernambuco, Recife 50670-901, PE, Brazil
| | - Luiz Carlos Alves
- Laboratory of Immunopathology Keizo Asami (LIKA) and Aggeu Magalhães Research Center, CPqAM/FIOCRUZ, Federal University of Pernambuco, Av. Moraes Rego s/n, Cidade Universitária, Recife 50670-420, PE, Brazil
| | - Fábio André Brayner dos Santos
- Laboratory of Immunopathology Keizo Asami (LIKA) and Aggeu Magalhães Research Center, CPqAM/FIOCRUZ, Federal University of Pernambuco, Av. Moraes Rego s/n, Cidade Universitária, Recife 50670-420, PE, Brazil
| | - Lucas Tricarico Barcellos
- Rural Federal University of Rio de Janeiro, Institute of Chemistry, Seropédica 23970-000, RJ, Brazil
| | | | - João Xavier de Araújo-Júnior
- Rural Federal University of Rio de Janeiro, Institute of Chemistry, Seropédica 23970-000, RJ, Brazil
- Laboratory of Medicinal Chemistry, Pharmaceutical Sciences Institute, Federal University of Alagoas, Campus A.C. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, Brazil
| | | | | | - Magna Suzana Alexandre-Moreira
- Laboratory of Pharmacology and Immunity, Institute of Biological Sciences and Health, Federal University of Alagoas, Campus A.C. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, AL, Brazil
| |
Collapse
|
6
|
Khalifa MM, Martorelli Di Genova B, McAlpine SG, Gallego-Lopez GM, Stevenson DM, Rozema SD, Monaghan NP, Morris JC, Knoll LJ, Golden JE. Dual-Stage Picolinic Acid-Derived Inhibitors of Toxoplasma gondii. ACS Med Chem Lett 2020; 11:2382-2388. [PMID: 33335660 DOI: 10.1021/acsmedchemlett.0c00267] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/07/2020] [Indexed: 12/20/2022] Open
Abstract
Toxoplasma gondii causes a prevalent human infection for which only the acute stage has an FDA-approved therapy. To find inhibitors of both the acute stage parasites and the persistent cyst stage that causes a chronic infection, we repurposed a compound library containing known inhibitors of parasitic hexokinase, the first step in the glycolysis pathway, along with a larger collection of new structural derivatives. The focused screen of 22 compounds showed a 77% hit rate (>50% multistage inhibition) and revealed a series of aminobenzamide-linked picolinic acids with submicromolar potency against both T. gondii parasite forms. Picolinic acid 23, designed from an antiparasitic benzamidobenzoic acid class with challenging ADME properties, showed 60-fold-enhanced solubility, a moderate LogD7.4, and a 30% improvement in microsomal stability. Furthermore, isotopically labeled glucose tracing revealed that picolinic acid 23 does not function by hexokinase inhibition. Thus, we report a new probe scaffold to interrogate dual-stage inhibition of T. gondii.
Collapse
Affiliation(s)
- Muhammad M. Khalifa
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, Wisconsin 53703, United States
| | - Bruno Martorelli Di Genova
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, 1550 Linden Drive, Madison, Wisconsin 53706, United States
| | - Sarah G. McAlpine
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, 190 Collins Street, Clemson, South Carolina 29634, United States
| | - Gina M. Gallego-Lopez
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, 1550 Linden Drive, Madison, Wisconsin 53706, United States
- Morgridge Institute for Research, Madison, Wisconsin 53715, United States
| | - David M. Stevenson
- Department of Bacteriology, University of Wisconsin-Madison, 1550 Linden Drive, Madison, Wisconsin 53706, United States of America
| | - Soren D. Rozema
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, Wisconsin 53703, United States
| | - Neil P. Monaghan
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, 190 Collins Street, Clemson, South Carolina 29634, United States
| | - James C. Morris
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, 190 Collins Street, Clemson, South Carolina 29634, United States
| | - Laura J. Knoll
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, 1550 Linden Drive, Madison, Wisconsin 53706, United States
| | - Jennifer E. Golden
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, Wisconsin 53703, United States
| |
Collapse
|
7
|
Garcia SN, Guedes RC, Marques MM. Unlocking the Potential of HK2 in Cancer Metabolism and Therapeutics. Curr Med Chem 2020; 26:7285-7322. [PMID: 30543165 DOI: 10.2174/0929867326666181213092652] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/26/2018] [Accepted: 11/06/2018] [Indexed: 12/24/2022]
Abstract
Glycolysis is a tightly regulated process in which several enzymes, such as Hexokinases (HKs), play crucial roles. Cancer cells are characterized by specific expression levels of several isoenzymes in different metabolic pathways and these features offer possibilities for therapeutic interventions. Overexpression of HKs (mostly of the HK2 isoform) have been consistently reported in numerous types of cancer. Moreover, deletion of HK2 has been shown to decrease cancer cell proliferation without explicit side effects in animal models, which suggests that targeting HK2 is a viable strategy for cancer therapy. HK2 inhibition causes a substantial decrease of glycolysis that affects multiple pathways of central metabolism and also destabilizes the mitochondrial outer membrane, ultimately enhancing cell death. Although glycolysis inhibition has met limited success, partly due to low selectivity for specific isoforms and excessive side effects of the reported HK inhibitors, there is ample ground for progress. The current review is focused on HK2 inhibition, envisaging the development of potent and selective anticancer agents. The information on function, expression, and activity of HKs is presented, along with their structures, known inhibitors, and reported effects of HK2 ablation/inhibition. The structural features of the different isozymes are discussed, aiming to stimulate a more rational approach to the design of selective HK2 inhibitors with appropriate drug-like properties. Particular attention is dedicated to a structural and sequence comparison of the structurally similar HK1 and HK2 isoforms, aiming to unveil differences that could be explored therapeutically. Finally, several additional catalytic- and non-catalytic roles on different pathways and diseases, recently attributed to HK2, are reviewed and their implications briefly discussed.
Collapse
Affiliation(s)
- Sara N Garcia
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal.,iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Rita C Guedes
- iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - M Matilde Marques
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| |
Collapse
|
8
|
Zak KM, Kalińska M, Wątor E, Kuśka K, Krutyhołowa R, Dubin G, Popowicz GM, Grudnik P. Crystal Structure of Kluyveromyces lactis Glucokinase ( KlGlk1). Int J Mol Sci 2019; 20:ijms20194821. [PMID: 31569356 PMCID: PMC6801647 DOI: 10.3390/ijms20194821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 11/30/2022] Open
Abstract
Glucose phosphorylating enzymes are crucial in the regulation of basic cellular processes, including metabolism and gene expression. Glucokinases and hexokinases provide a pool of phosphorylated glucose in an adenosine diphosphate (ADP)- and ATP-dependent manner to shape the cell metabolism. The glucose processing enzymes from Kluyveromyces lactis are poorly characterized despite the emerging contribution of this yeast strain to industrial and laboratory scale biotechnology. The first reports on K. lactis glucokinase (KlGlk1) positioned the enzyme as an essential component required for glucose signaling. Nevertheless, no biochemical and structural information was available until now. Here, we present the first crystal structure of KlGlk1 together with biochemical characterization, including substrate specificity and enzyme kinetics. Additionally, comparative analysis of the presented structure and the prior structures of lactis hexokinase (KlHxk1) demonstrates the potential transitions between open and closed enzyme conformations upon ligand binding.
Collapse
Affiliation(s)
- Krzysztof M Zak
- Institute of Structural Biology, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
- Malopolska Centre of Biotechnology, Jagiellonian University, ul. Gronostajowa 7a, 30-387 Krakow, Poland.
| | - Magdalena Kalińska
- Malopolska Centre of Biotechnology, Jagiellonian University, ul. Gronostajowa 7a, 30-387 Krakow, Poland.
| | - Elżbieta Wątor
- Malopolska Centre of Biotechnology, Jagiellonian University, ul. Gronostajowa 7a, 30-387 Krakow, Poland.
| | - Katarzyna Kuśka
- Malopolska Centre of Biotechnology, Jagiellonian University, ul. Gronostajowa 7a, 30-387 Krakow, Poland.
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Krakow, Poland.
| | - Rościsław Krutyhołowa
- Malopolska Centre of Biotechnology, Jagiellonian University, ul. Gronostajowa 7a, 30-387 Krakow, Poland.
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Krakow, Poland.
| | - Grzegorz Dubin
- Malopolska Centre of Biotechnology, Jagiellonian University, ul. Gronostajowa 7a, 30-387 Krakow, Poland.
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Krakow, Poland.
| | - Grzegorz M Popowicz
- Institute of Structural Biology, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
- Center for Integrated Protein Science Munich at Chair of Biomolecular NMR, Department Chemie, Technische Universität München, Lichtenbergstrasse 4, 85747 Garching, Germany.
| | - Przemysław Grudnik
- Malopolska Centre of Biotechnology, Jagiellonian University, ul. Gronostajowa 7a, 30-387 Krakow, Poland.
| |
Collapse
|
9
|
Enzymatic and Structural Characterization of the Naegleria fowleri Glucokinase. Antimicrob Agents Chemother 2019; 63:AAC.02410-18. [PMID: 30783001 DOI: 10.1128/aac.02410-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/08/2019] [Indexed: 02/07/2023] Open
Abstract
Infection with the free-living amoeba Naegleria fowleri leads to life-threatening primary amoebic meningoencephalitis. Efficacious treatment options for these infections are limited, and the mortality rate is very high (∼98%). Parasite metabolism may provide suitable targets for therapeutic design. Like most other organisms, glucose metabolism is critical for parasite viability, being required for growth in culture. The first enzyme required for glucose metabolism is typically a hexokinase (HK), which transfers a phosphate from ATP to glucose. The products of this enzyme are required for both glycolysis and the pentose phosphate pathway. However, the N. fowleri genome lacks an obvious HK homolog and instead harbors a glucokinase (Glck). The N. fowleri Glck (NfGlck) shares limited (25%) amino acid identity with the mammalian host enzyme (Homo sapiens Glck), suggesting that parasite-specific inhibitors with anti-amoeba activity can be generated. Following heterologous expression, NfGlck was found to have a limited hexose substrate range, with the greatest activity observed with glucose. The enzyme had apparent Km values of 42.5 ± 7.3 μM and 141.6 ± 9.9 μM for glucose and ATP, respectively. The NfGlck structure was determined and refined to 2.2-Å resolution, revealing that the enzyme shares greatest structural similarity with the Trypanosoma cruzi Glck. These similarities include binding modes and binding environments for substrates. To identify inhibitors of NfGlck, we screened a small collection of inhibitors of glucose-phosphorylating enzymes and identified several small molecules with 50% inhibitory concentration values of <1 μM that may prove useful as hit chemotypes for further leads and therapeutic development against N. fowleri.
Collapse
|
10
|
Patel OPS, Dhiman S, Khan S, Shinde VN, Jaspal S, Srivathsa MR, Jha PN, Kumar A. A straightforward TBHP-mediated synthesis of 2-amidobenzoic acids from 2-arylindoles and their antimicrobial activity. Org Biomol Chem 2019; 17:5962-5970. [DOI: 10.1039/c9ob00797k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Synthesis of 2-amidobenzoic acids has been achieved through TBHP-mediated oxidative ring opening of 2-arylindoles. The synthesized compounds have been evaluated for their antimicrobial activity.
Collapse
Affiliation(s)
| | - Shiv Dhiman
- Department of Chemistry
- BITS Pilani
- Pilani Campus
- India
| | - Shahid Khan
- Department of Biological Sciences
- BITS Pilani
- Pilani Campus
- India
| | | | - Sonam Jaspal
- Department of Chemistry
- BITS Pilani
- Pilani Campus
- India
| | | | - Prabhat N. Jha
- Department of Biological Sciences
- BITS Pilani
- Pilani Campus
- India
| | - Anil Kumar
- Department of Chemistry
- BITS Pilani
- Pilani Campus
- India
| |
Collapse
|
11
|
Kalel VC, Mäser P, Sattler M, Erdmann R, Popowicz GM. Come, sweet death: targeting glycosomal protein import for antitrypanosomal drug development. Curr Opin Microbiol 2018; 46:116-122. [PMID: 30481613 DOI: 10.1016/j.mib.2018.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/09/2018] [Indexed: 01/18/2023]
Abstract
Glycosomes evolved as specialized system for glycolysis in trypanosomatids. These organelle rely on protein import to maintain function. A machinery of peroxin (PEX) proteins is responsible for recognition and transport of glycosomal proteins to the organelle. Disruption of PEX-based import system was expected to be a strategy against trypanosomatids. Recently, a proof of this hypothesis has been presented. Here, we review current information about trypanosomatids' glycosomal transport components as targets for new trypanocidal therapies.
Collapse
Affiliation(s)
- Vishal C Kalel
- Institute of Biochemistry and Pathobiochemistry, Department of Systems Biochemistry, Faculty of Medicine, Ruhr University Bochum, 44780 Bochum, Germany
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4051 Basel, Switzerland; University of Basel, 4001 Basel, Switzerland
| | - Michael Sattler
- Institute of Structural Biology, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Center for Integrated Protein Science Munich at Chair of Biomolecular NMR, Department Chemie, Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany
| | - Ralf Erdmann
- Institute of Biochemistry and Pathobiochemistry, Department of Systems Biochemistry, Faculty of Medicine, Ruhr University Bochum, 44780 Bochum, Germany
| | - Grzegorz M Popowicz
- Institute of Structural Biology, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Center for Integrated Protein Science Munich at Chair of Biomolecular NMR, Department Chemie, Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany.
| |
Collapse
|
12
|
Voyton CM, Qiu Y, Morris MT, Ackroyd PC, Suryadi J, Crowe L, Morris JC, Christensen KA. A FRET flow cytometry method for monitoring cytosolic and glycosomal glucose in living kinetoplastid parasites. PLoS Negl Trop Dis 2018; 12:e0006523. [PMID: 29851949 PMCID: PMC5997345 DOI: 10.1371/journal.pntd.0006523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/12/2018] [Accepted: 05/11/2018] [Indexed: 11/18/2022] Open
Abstract
The bloodstream lifecycle stage of the kinetoplastid parasite Trypanosoma brucei relies solely on glucose metabolism for ATP production, which occurs in peroxisome-like organelles (glycosomes). Many studies have been conducted on glucose uptake and metabolism, but none thus far have been able to monitor changes in cellular and organellar glucose concentration in live parasites. We have developed a non-destructive technique for monitoring changes in cytosolic and glycosomal glucose levels in T. brucei using a fluorescent protein biosensor (FLII12Pglu-700μδ6) in combination with flow cytometry. T. brucei parasites harboring the biosensor allowed for observation of cytosolic glucose levels. Appending a type 1 peroxisomal targeting sequence caused biosensors to localize to glycosomes, which enabled observation of glycosomal glucose levels. Using this approach, we investigated cytosolic and glycosomal glucose levels in response to changes in external glucose or 2-deoxyglucose concentration. These data show that procyclic form and bloodstream form parasites maintain different glucose concentrations in their cytosol and glycosomes. In procyclic form parasites, the cytosol and glycosomes maintain indistinguishable glucose levels (3.4 ± 0.4mM and 3.4 ± 0.5mM glucose respectively) at a 6.25mM external glucose concentration. In contrast, bloodstream form parasites maintain glycosomal glucose levels that are ~1.8-fold higher than the surrounding cytosol, equating to 1.9 ± 0.6mM in cytosol and 3.5 ± 0.5mM in glycosomes. While the mechanisms of glucose transport operating in the glycosomes of bloodstream form T. brucei remain unresolved, the methods described here will provide a means to begin to dissect the cellular machinery required for subcellular distribution of this critical hexose. African sleeping sickness is caused by Trypanosoma brucei. Tens of millions of people living in endemic areas are at risk for the disease. Within the mammalian bloodstream, T. brucei parasites sustain all their energy needs by metabolizing glucose present in the host’s blood within specialized organelles known as glycosomes. In vitro, bloodstream parasites rapidly die if glucose is removed from their environment. This reliance on glucose for survival has made glucose metabolism in T. brucei an important area of study with the aim to develop targeted therapeutics that disrupt glucose metabolism. However, there have previously been no reported methods to study glucose uptake and distribution dynamics in intact glycosomes in live T. brucei. Here we describe development of approaches for observing changes in glucose concentration in glycosomes in live T. brucei. Results obtained using these methods provide new insights into how T. brucei acquires and transports glucose to sustain cell survival.
Collapse
Affiliation(s)
- Charles M. Voyton
- Department of Chemistry, Clemson University, Clemson, South Carolina, United States of America
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Yijian Qiu
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, United States of America
| | - Meredith T. Morris
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, United States of America
| | - P. Christine Ackroyd
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Jimmy Suryadi
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, United States of America
| | - Logan Crowe
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, United States of America
| | - James C. Morris
- Eukaryotic Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, United States of America
| | - Kenneth A. Christensen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
- * E-mail:
| |
Collapse
|