1
|
Ober VT, Githure GB, Volpato Santos Y, Becker S, Moya Munoz G, Basquin J, Schwede F, Lorentzen E, Boshart M. Purine nucleosides replace cAMP in allosteric regulation of PKA in trypanosomatid pathogens. eLife 2024; 12:RP91040. [PMID: 38517938 PMCID: PMC10959531 DOI: 10.7554/elife.91040] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024] Open
Abstract
Cyclic nucleotide binding domains (CNB) confer allosteric regulation by cAMP or cGMP to many signaling proteins, including PKA and PKG. PKA of phylogenetically distant Trypanosoma is the first exception as it is cyclic nucleotide-independent and responsive to nucleoside analogues (Bachmaier et al., 2019). Here, we show that natural nucleosides inosine, guanosine and adenosine are nanomolar affinity CNB ligands and activators of PKA orthologs of the important tropical pathogens Trypanosoma brucei, Trypanosoma cruzi, and Leishmania. The sequence and structural determinants of binding affinity, -specificity and kinase activation of PKAR were established by structure-activity relationship (SAR) analysis, co-crystal structures and mutagenesis. Substitution of two to three amino acids in the binding sites is sufficient for conversion of CNB domains from nucleoside to cyclic nucleotide specificity. In addition, a trypanosomatid-specific C-terminal helix (αD) is required for high affinity binding to CNB-B. The αD helix functions as a lid of the binding site that shields ligands from solvent. Selectivity of guanosine for CNB-B and of adenosine for CNB-A results in synergistic kinase activation at low nanomolar concentration. PKA pulldown from rapid lysis establishes guanosine as the predominant ligand in vivo in T. brucei bloodstream forms, whereas guanosine and adenosine seem to synergize in the procyclic developmental stage in the insect vector. We discuss the versatile use of CNB domains in evolution and recruitment of PKA for novel nucleoside-mediated signaling.
Collapse
Affiliation(s)
- Veronica Teresa Ober
- Faculty of Biology, Genetics, Ludwig-Maximilians University Munich (LMU)MartinsriedGermany
| | | | - Yuri Volpato Santos
- Faculty of Biology, Genetics, Ludwig-Maximilians University Munich (LMU)MartinsriedGermany
| | - Sidney Becker
- Max Planck Institute of Molecular PhysiologyDortmundGermany
- TU Dortmund, Department of Chemistry and Chemical BiologyDortmundGermany
| | - Gabriel Moya Munoz
- Faculty of Biology, Genetics, Ludwig-Maximilians University Munich (LMU)MartinsriedGermany
| | | | - Frank Schwede
- BIOLOG Life Science Institute GmbH & Co KGBremenGermany
| | - Esben Lorentzen
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Michael Boshart
- Faculty of Biology, Genetics, Ludwig-Maximilians University Munich (LMU)MartinsriedGermany
| |
Collapse
|
2
|
Kaiser A, Agostinelli E. Hypusinated EIF5A as a feasible drug target for Advanced Medicinal Therapies in the treatment of pathogenic parasites and therapy-resistant tumors. Amino Acids 2022; 54:501-511. [DOI: 10.1007/s00726-021-03120-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/21/2021] [Indexed: 11/30/2022]
|
3
|
Kaiser A, Doerig C. Editorial: Heading Against Parasitic Resistance: A Screen for Next Generation Drugs Against Targets of cAMP- or cGMP-regulated Pathways. Front Microbiol 2021; 12:727978. [PMID: 34484170 PMCID: PMC8415706 DOI: 10.3389/fmicb.2021.727978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 07/22/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Annette Kaiser
- Medical Research Centre, University of Duisburg-Essen, Duisburg, Germany
| | - Christian Doerig
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
4
|
Lasonder E, More K, Singh S, Haidar M, Bertinetti D, Kennedy EJ, Herberg FW, Holder AA, Langsley G, Chitnis CE. cAMP-Dependent Signaling Pathways as Potential Targets for Inhibition of Plasmodium falciparum Blood Stages. Front Microbiol 2021; 12:684005. [PMID: 34108954 PMCID: PMC8183823 DOI: 10.3389/fmicb.2021.684005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/30/2021] [Indexed: 11/13/2022] Open
Abstract
We review the role of signaling pathways in regulation of the key processes of merozoite egress and red blood cell invasion by Plasmodium falciparum and, in particular, the importance of the second messengers, cAMP and Ca2+, and cyclic nucleotide dependent kinases. cAMP-dependent protein kinase (PKA) is comprised of cAMP-binding regulatory, and catalytic subunits. The less well conserved cAMP-binding pockets should make cAMP analogs attractive drug leads, but this approach is compromised by the poor membrane permeability of cyclic nucleotides. We discuss how the conserved nature of ATP-binding pockets makes ATP analogs inherently prone to off-target effects and how ATP analogs and genetic manipulation can be useful research tools to examine this. We suggest that targeting PKA interaction partners as well as substrates, or developing inhibitors based on PKA interaction sites or phosphorylation sites in PKA substrates, may provide viable alternative approaches for the development of anti-malarial drugs. Proximity of PKA to a substrate is necessary for substrate phosphorylation, but the P. falciparum genome encodes few recognizable A-kinase anchor proteins (AKAPs), suggesting the importance of PKA-regulatory subunit myristylation and membrane association in determining substrate preference. We also discuss how Pf14-3-3 assembles a phosphorylation-dependent signaling complex that includes PKA and calcium dependent protein kinase 1 (CDPK1) and how this complex may be critical for merozoite invasion, and a target to block parasite growth. We compare altered phosphorylation levels in intracellular and egressed merozoites to identify potential PKA substrates. Finally, as host PKA may have a critical role in supporting intracellular parasite development, we discuss its role at other stages of the life cycle, as well as in other apicomplexan infections. Throughout our review we propose possible new directions for the therapeutic exploitation of cAMP-PKA-signaling in malaria and other diseases caused by apicomplexan parasites.
Collapse
Affiliation(s)
- Edwin Lasonder
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Kunal More
- Unité de Biologie de Plasmodium et Vaccins, Département de Parasites et Insectes Vecteurs, Institut Pasteur, Paris, France
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Malak Haidar
- Laboratoire de Biologie Comparative des Apicomplexes, Faculté de Médecine, Université Paris Descartes - Sorbonne Paris Cité, Paris, France.,INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France
| | | | - Eileen J Kennedy
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, United States
| | | | - Anthony A Holder
- Malaria Parasitology Laboratory, Francis Crick Institute, London, United Kingdom
| | - Gordon Langsley
- Laboratoire de Biologie Comparative des Apicomplexes, Faculté de Médecine, Université Paris Descartes - Sorbonne Paris Cité, Paris, France.,INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France
| | - Chetan E Chitnis
- Unité de Biologie de Plasmodium et Vaccins, Département de Parasites et Insectes Vecteurs, Institut Pasteur, Paris, France
| |
Collapse
|
5
|
Bardeci NG, Tofolón E, Trajtenberg F, Caramelo J, Larrieux N, Rossi S, Buschiazzo A, Moreno S. The crystal structure of yeast regulatory subunit reveals key evolutionary insights into Protein Kinase A oligomerization. J Struct Biol 2021; 213:107732. [PMID: 33819633 DOI: 10.1016/j.jsb.2021.107732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/15/2021] [Accepted: 03/30/2021] [Indexed: 02/08/2023]
Abstract
Protein Kinase A (PKA) is a widespread enzyme that plays a key role in many signaling pathways from lower eukaryotes to metazoans. In mammals, the regulatory (R) subunits sequester and target the catalytic (C) subunits to proper subcellular locations. This targeting is accomplished by the dimerization and docking (D/D) domain of the R subunits. The activation of the holoenzyme depends on the binding of the second messenger cAMP. The only available structures of the D/D domain proceed from mammalian sources. Unlike dimeric mammalian counterparts, the R subunit from Saccharomyces cerevisiae (Bcy1) forms tetramers in solution. Here we describe the first high-resolution structure of a non-mammalian D/D domain. The tetramer in the crystals of the Bcy1 D/D domain is a dimer of dimers that retain the classical D/D domain fold. By using phylogenetic and structural analyses combined with site-directed mutagenesis, we found that fungal R subunits present an insertion of a single amino acid at the D/D domain that shifts the position of a downstream, conserved arginine. This residue participates in intra-dimer interactions in mammalian D/D domains, while due to this insertion it is involved in inter-dimer contacts in Bcy1, which are crucial for the stability of the tetramer. This surprising finding challenges well-established concepts regarding the oligomeric state within the PKAR protein family and provides important insights into the yet unexplored structural diversity of the D/D domains and the molecular determinants of R subunit oligomerization.
Collapse
Affiliation(s)
- Nicolás González Bardeci
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; Instituto de Química Biológica, Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires C1428EHA, Argentina.
| | - Enzo Tofolón
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; Instituto de Química Biológica, Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires C1428EHA, Argentina
| | - Felipe Trajtenberg
- Laboratory of Molecular and Structural Microbiology, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Julio Caramelo
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; Fundación Instituto Leloir, Instituto de investigaciones Bioquímicas de Buenos Aires (IIBBA- CONICET), Buenos Aires C1405BWE, Argentina
| | - Nicole Larrieux
- Laboratory of Molecular and Structural Microbiology, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Silvia Rossi
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; Instituto de Química Biológica, Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires C1428EHA, Argentina
| | - Alejandro Buschiazzo
- Laboratory of Molecular and Structural Microbiology, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Silvia Moreno
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; Instituto de Química Biológica, Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires C1428EHA, Argentina.
| |
Collapse
|
6
|
Moolman C, van der Sluis R, Beteck RM, Legoabe LJ. An Update on Development of Small-Molecule Plasmodial Kinase Inhibitors. Molecules 2020; 25:E5182. [PMID: 33171706 PMCID: PMC7664427 DOI: 10.3390/molecules25215182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/21/2022] Open
Abstract
Malaria control relies heavily on the small number of existing antimalarial drugs. However, recurring antimalarial drug resistance necessitates the continual generation of new antimalarial drugs with novel modes of action. In order to shift the focus from only controlling this disease towards elimination and eradication, next-generation antimalarial agents need to address the gaps in the malaria drug arsenal. This includes developing drugs for chemoprotection, treating severe malaria and blocking transmission. Plasmodial kinases are promising targets for next-generation antimalarial drug development as they mediate critical cellular processes and some are active across multiple stages of the parasite's life cycle. This review gives an update on the progress made thus far with regards to plasmodial kinase small-molecule inhibitor development.
Collapse
Affiliation(s)
- Chantalle Moolman
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa; (C.M.); (R.M.B.)
| | - Rencia van der Sluis
- Focus Area for Human Metabolomics, Biochemistry, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa;
| | - Richard M. Beteck
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa; (C.M.); (R.M.B.)
| | - Lesetja J. Legoabe
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa; (C.M.); (R.M.B.)
| |
Collapse
|
7
|
Ji S, Xiong Y, Lu W, Li M, Wang X, Wang C, Wang D, Xiao J, Zhu Z, Chen L, Zhang Y, Qing G. cAMP sensitive nanochannels driven by conformational transition of a tripeptide-based smart polymer. Chem Commun (Camb) 2020; 56:3425-3428. [PMID: 32100737 DOI: 10.1039/c9cc09588h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Inspired by biological nanochannels, a novel cyclic 3',5'-adenosine monophosphate (cAMP)-regulated artificial nanochannel based on a tripeptide Arg-Thr-Ala (RTA) design is developed. Highly specific binding between the tripeptide and cAMP triggers an obvious conformational transition of a smart polymer chain from a contracted state to a swollen one, which leads to a dynamic modulation of the gating behaviours of the nanochannels.
Collapse
Affiliation(s)
- Shengyan Ji
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, P. R. China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Abstract
Understanding the mechanisms behind host cell invasion by Plasmodium falciparum remains a major hurdle to developing antimalarial therapeutics that target the asexual cycle and the symptomatic stage of malaria. Host cell entry is enabled by a multitude of precisely timed and tightly regulated receptor-ligand interactions. Cyclic nucleotide signaling has been implicated in regulating parasite invasion, and an important downstream effector of the cAMP-signaling pathway is protein kinase A (PKA), a cAMP-dependent protein kinase. There is increasing evidence that P. falciparum PKA (PfPKA) is responsible for phosphorylation of the cytoplasmic domain of P. falciparum apical membrane antigen 1 (PfAMA1) at Ser610, a cAMP-dependent event that is crucial for successful parasite invasion. In the present study, CRISPR-Cas9 and conditional gene deletion (dimerizable cre) technologies were implemented to generate a P. falciparum parasite line in which expression of the catalytic subunit of PfPKA (PfPKAc) is under conditional control, demonstrating highly efficient dimerizable Cre recombinase (DiCre)-mediated gene excision and complete knockdown of protein expression. Parasites lacking PfPKAc show severely reduced growth after one intraerythrocytic growth cycle and are deficient in host cell invasion, as highlighted by live-imaging experiments. Furthermore, PfPKAc-deficient parasites are unable to phosphorylate PfAMA1 at Ser610. This work not only identifies an essential role for PfPKAc in the P. falciparum asexual life cycle but also confirms that PfPKAc is the kinase responsible for phosphorylating PfAMA1 Ser610.IMPORTANCE Malaria continues to present a major global health burden, particularly in low-resource countries. Plasmodium falciparum, the parasite responsible for the most severe form of malaria, causes disease through rapid and repeated rounds of invasion and replication within red blood cells. Invasion into red blood cells is essential for P. falciparum survival, and the molecular events mediating this process have gained much attention as potential therapeutic targets. With no effective vaccine available, and with the emergence of resistance to antimalarials, there is an urgent need for the development of new therapeutics. Our research has used genetic techniques to provide evidence of an essential protein kinase involved in P. falciparum invasion. Our work adds to the current understanding of parasite signaling processes required for invasion, highlighting PKA as a potential drug target to inhibit invasion for the treatment of malaria.
Collapse
|
9
|
Gilson PR, Kumarasingha R, Thompson J, Zhang X, Penington JS, Kalhor R, Bullen HE, Lehane AM, Dans MG, de Koning-Ward TF, Holien JK, Soares da Costa TP, Hulett MD, Buskes MJ, Crabb BS, Kirk K, Papenfuss AT, Cowman AF, Abbott BM. A 4-cyano-3-methylisoquinoline inhibitor of Plasmodium falciparum growth targets the sodium efflux pump PfATP4. Sci Rep 2019; 9:10292. [PMID: 31311978 PMCID: PMC6635429 DOI: 10.1038/s41598-019-46500-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/28/2019] [Indexed: 12/31/2022] Open
Abstract
We developed a novel series of antimalarial compounds based on a 4-cyano-3-methylisoquinoline. Our lead compound MB14 achieved modest inhibition of the growth in vitro of the human malaria parasite, Plasmodium falciparum. To identify its biological target we selected for parasites resistant to MB14. Genome sequencing revealed that all resistant parasites bore a single point S374R mutation in the sodium (Na+) efflux transporter PfATP4. There are many compounds known to inhibit PfATP4 and some are under preclinical development. MB14 was shown to inhibit Na+ dependent ATPase activity in parasite membranes, consistent with the compound targeting PfATP4 directly. PfATP4 inhibitors cause swelling and lysis of infected erythrocytes, attributed to the accumulation of Na+ inside the intracellular parasites and the resultant parasite swelling. We show here that inhibitor-induced lysis of infected erythrocytes is dependent upon the parasite protein RhopH2, a component of the new permeability pathways that are induced by the parasite in the erythrocyte membrane. These pathways mediate the influx of Na+ into the infected erythrocyte and their suppression via RhopH2 knockdown limits the accumulation of Na+ within the parasite hence protecting the infected erythrocyte from lysis. This study reveals a role for the parasite-induced new permeability pathways in the mechanism of action of PfATP4 inhibitors.
Collapse
Affiliation(s)
- Paul R Gilson
- Burnet Institute, Melbourne, Victoria, 3004, Australia. .,Monash University, Melbourne, Victoria, 3800, Australia.
| | | | - Jennifer Thompson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
| | - Xinxin Zhang
- Research School of Biology, Australian National University, Canberra, ACT, 2601, Australia
| | | | - Robabeh Kalhor
- La Trobe University, Melbourne, Victoria, 3086, Australia
| | | | - Adele M Lehane
- Research School of Biology, Australian National University, Canberra, ACT, 2601, Australia
| | - Madeline G Dans
- Burnet Institute, Melbourne, Victoria, 3004, Australia.,School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | | | - Jessica K Holien
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, 3065, Australia
| | | | - Mark D Hulett
- La Trobe University, Melbourne, Victoria, 3086, Australia
| | | | - Brendan S Crabb
- Burnet Institute, Melbourne, Victoria, 3004, Australia.,Monash University, Melbourne, Victoria, 3800, Australia.,University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Kiaran Kirk
- Research School of Biology, Australian National University, Canberra, ACT, 2601, Australia
| | - Anthony T Papenfuss
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.,University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Alan F Cowman
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.,University of Melbourne, Melbourne, Victoria, 3010, Australia
| | | |
Collapse
|
10
|
Baker DA, Drought LG, Flueck C, Nofal SD, Patel A, Penzo M, Walker EM. Cyclic nucleotide signalling in malaria parasites. Open Biol 2018; 7:rsob.170213. [PMID: 29263246 PMCID: PMC5746546 DOI: 10.1098/rsob.170213] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/28/2017] [Indexed: 12/22/2022] Open
Abstract
The cyclic nucleotides 3′, 5′-cyclic adenosine monophosphate (cAMP) and 3′, 5′-cyclic guanosine monophosphate (cGMP) are intracellular messengers found in most animal cell types. They usually mediate an extracellular stimulus to drive a change in cell function through activation of their respective cyclic nucleotide-dependent protein kinases, PKA and PKG. The enzymatic components of the malaria parasite cyclic nucleotide signalling pathways have been identified, and the genetic and biochemical studies of these enzymes carried out to date are reviewed herein. What has become very clear is that cyclic nucleotides play vital roles in controlling every stage of the complex malaria parasite life cycle. Our understanding of the involvement of cyclic nucleotide signalling in orchestrating the complex biology of malaria parasites is still in its infancy, but the recent advances in our genetic tools and the increasing interest in signalling will deliver more rapid progress in the coming years.
Collapse
Affiliation(s)
- David A Baker
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Laura G Drought
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Christian Flueck
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Stephanie D Nofal
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Avnish Patel
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Maria Penzo
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK.,Tres Cantos Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos, 28760, Madrid, Spain
| | - Eloise M Walker
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
11
|
Cabrera DG, Horatscheck A, Wilson CR, Basarab G, Eyermann CJ, Chibale K. Plasmodial Kinase Inhibitors: License to Cure? J Med Chem 2018; 61:8061-8077. [PMID: 29771541 PMCID: PMC6166223 DOI: 10.1021/acs.jmedchem.8b00329] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
Advances
in the genetics, function, and stage-specificity of Plasmodium kinases has driven robust efforts to identify
targets for the design of antimalarial therapies. Reverse genomics
following phenotypic screening against Plasmodia or
related parasites has uncovered vulnerable kinase targets including
PI4K, PKG, and GSK-3, an approach bolstered by access to human disease-directed
kinase libraries. Alternatively, screening compound libraries against Plasmodium kinases has successfully led to inhibitors with
antiplasmodial activity. As with other therapeutic areas, optimizing
compound ADMET and PK properties in parallel with target inhibitory
potency and whole cell activity becomes paramount toward advancing
compounds as clinical candidates. These and other considerations will
be discussed in the context of progress achieved toward deriving important,
novel mode-of-action kinase-inhibiting antimalarial medicines.
Collapse
|
12
|
Jia Y, Marq JB, Bisio H, Jacot D, Mueller C, Yu L, Choudhary J, Brochet M, Soldati-Favre D. Crosstalk between PKA and PKG controls pH-dependent host cell egress of Toxoplasma gondii. EMBO J 2017; 36:3250-3267. [PMID: 29030485 PMCID: PMC5666616 DOI: 10.15252/embj.201796794] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 09/08/2017] [Accepted: 09/12/2017] [Indexed: 12/27/2022] Open
Abstract
Toxoplasma gondii encodes three protein kinase A catalytic (PKAc1-3) and one regulatory (PKAr) subunits to integrate cAMP-dependent signals. Here, we show that inactive PKAc1 is maintained at the parasite pellicle by interacting with acylated PKAr. Either a conditional knockdown of PKAr or the overexpression of PKAc1 blocks parasite division. Conversely, down-regulation of PKAc1 or stabilisation of a dominant-negative PKAr isoform that does not bind cAMP triggers premature parasite egress from infected cells followed by serial invasion attempts leading to host cell lysis. This untimely egress depends on host cell acidification. A phosphoproteome analysis suggested the interplay between cAMP and cGMP signalling as PKAc1 inactivation changes the phosphorylation profile of a putative cGMP-phosphodiesterase. Concordantly, inhibition of the cGMP-dependent protein kinase G (PKG) blocks egress induced by PKAc1 inactivation or environmental acidification, while a cGMP-phosphodiesterase inhibitor circumvents egress repression by PKAc1 or pH neutralisation. This indicates that pH and PKAc1 act as balancing regulators of cGMP metabolism to control egress. These results reveal a crosstalk between PKA and PKG pathways to govern egress in T. gondii.
Collapse
Affiliation(s)
- Yonggen Jia
- Department of Microbiology and Molecular Medicine, CMU, University of Geneva, Geneva 4, Switzerland
| | - Jean-Baptiste Marq
- Department of Microbiology and Molecular Medicine, CMU, University of Geneva, Geneva 4, Switzerland
| | - Hugo Bisio
- Department of Microbiology and Molecular Medicine, CMU, University of Geneva, Geneva 4, Switzerland
| | - Damien Jacot
- Department of Microbiology and Molecular Medicine, CMU, University of Geneva, Geneva 4, Switzerland
| | - Christina Mueller
- Department of Microbiology and Molecular Medicine, CMU, University of Geneva, Geneva 4, Switzerland
| | - Lu Yu
- Proteomic Mass-spectrometry Team, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Jyoti Choudhary
- Proteomic Mass-spectrometry Team, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Mathieu Brochet
- Department of Microbiology and Molecular Medicine, CMU, University of Geneva, Geneva 4, Switzerland
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, CMU, University of Geneva, Geneva 4, Switzerland
| |
Collapse
|