1
|
Khim M, Montgomery J, Laureano De Souza M, Delvillar M, Weible LJ, Prabakaran M, Hulverson MA, Eck T, Bheemanabonia RY, Alday PH, Rotella DP, Doggett JS, Staker BL, Ojo KK, Bhanot P. Versatile Imidazole Scaffold with Potent Activity against Multiple Apicomplexan Parasites. ACS Infect Dis 2025. [PMID: 40339062 DOI: 10.1021/acsinfecdis.5c00049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Malaria, toxoplasmosis, and cryptosporidiosis are caused by apicomplexan parasites Plasmodium spp., Toxoplasma gondii, and Cryptosporidium parvum, respectively, and pose major health challenges. Their therapies are inadequate, ineffective or threatened by drug resistance. The development of novel drugs against them requires innovative and resource-efficient strategies. We exploited the kinome conservation of these parasites to determine the cellular targets and effects of two Plasmodium falciparum inhibitors in T. gondii and C. parvum. The imidazoles, (R)-RY-1-165 and (R)-RY-1-185, were developed to target the cGMP dependent protein kinase of P. falciparum (PfPKG), orthologs of which are present in T. gondii and C. parvum. Using structural and modeling approaches we determined that the molecules bind stereospecifically and interact with PfPKG in a manner unique among described inhibitors. We used enzymatic assays and mutant P. falciparum expressing PfPKG with a substituted "gatekeeper" residue to determine that cellular activity of the molecules is mediated through targets additional to PfPKG. These likely include P. falciparum calcium dependent protein kinase 1 and 4 (PfCDPK-1, -4), kinases that, like PfPKG, have small amino acids at the "gatekeeper" position. The molecules are active against T. gondii and C. parvum, with T. gondii tachyzoites being particularly sensitive. Using mutant parasites, enzyme assays and modeling studies we demonstrate that targets in T. gondii include TgPKG, TgCDPK1, TgCDPK4 and the mitogen activated kinase-like 1 (MAPKL-1). Our results suggest that this scaffold holds promise for the development of new toxoplasmosis drugs.
Collapse
Affiliation(s)
- Monique Khim
- Seattle Structural Genomics Center for Infectious Disease, Seattle, Washington 98109, United States
- Center for Global Infectious Disease ResearchSeattle Children's Research Institute, Seattle, Washington 98109, United States
| | - Jemma Montgomery
- Divisions of Infectious Diseases and ResearchVA Portland Healthcare System, Portland, Oregon 97239, United States
| | - Mariana Laureano De Souza
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Melvin Delvillar
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Lyssa J Weible
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious DiseasesUniversity of Washington, Seattle, Washington 98109, United States
| | - Mayuri Prabakaran
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious DiseasesUniversity of Washington, Seattle, Washington 98109, United States
| | - Matthew A Hulverson
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious DiseasesUniversity of Washington, Seattle, Washington 98109, United States
| | - Tyler Eck
- Department of Chemistry and Biochemistry and Sokol Institute of Pharmaceutical Life SciencesMontclair State University, Montclair, New Jersey 07043, United States
| | - Rammohan Y Bheemanabonia
- Department of Chemistry and Biochemistry and Sokol Institute of Pharmaceutical Life SciencesMontclair State University, Montclair, New Jersey 07043, United States
| | - P Holland Alday
- Divisions of Infectious Diseases and ResearchVA Portland Healthcare System, Portland, Oregon 97239, United States
- Division of Infectious Diseases,Oregon Health & Science University School of Medicine, Portland, Oregon 97239, United States
| | - David P Rotella
- Department of Chemistry and Biochemistry and Sokol Institute of Pharmaceutical Life SciencesMontclair State University, Montclair, New Jersey 07043, United States
| | - J Stone Doggett
- Divisions of Infectious Diseases and ResearchVA Portland Healthcare System, Portland, Oregon 97239, United States
- Division of Infectious Diseases,Oregon Health & Science University School of Medicine, Portland, Oregon 97239, United States
| | - Bart L Staker
- Seattle Structural Genomics Center for Infectious Disease, Seattle, Washington 98109, United States
- Center for Global Infectious Disease ResearchSeattle Children's Research Institute, Seattle, Washington 98109, United States
| | - Kayode K Ojo
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious DiseasesUniversity of Washington, Seattle, Washington 98109, United States
- Department of Global HealthUniversity of Washington, Seattle, Washington 98195, United States
| | - Purnima Bhanot
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey 07103, United States
| |
Collapse
|
2
|
Sánchez-Sánchez R, Huertas-López A, Largo-de la Torre A, Ferre I, Dini FM, Re M, Moreno-Gonzalo J, Choi R, Hulverson MA, Ojo KK, Arnold SLM, Hemphill A, Van Voorhis WC, Ortega-Mora LM. Treatment with BKI-1748 after Toxoplasma gondii systemic dissemination in experimentally infected pregnant sheep improves fetal and lamb mortality and morbidity and prevents congenital infection. Antimicrob Agents Chemother 2025; 69:e0144824. [PMID: 39745365 PMCID: PMC11823607 DOI: 10.1128/aac.01448-24] [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: 09/26/2024] [Accepted: 12/07/2024] [Indexed: 02/14/2025] Open
Abstract
Drug development for congenital toxoplasmosis is challenging since first-line therapy has a high rate of adverse effects and exhibits suboptimal efficacy. Bumped kinase inhibitors (BKIs), targeting protein kinases with small gatekeeper residues, have been found to be effective against Toxoplasma gondii. The efficacy of BKI-1748 administered later than 2 days post-infection (p.i.), a scenario that may better reflect its real-world use as a therapeutic candidate, has not been investigated in T. gondii-infected pregnant sheep. For this purpose, 19 pregnant sheep were assigned to three experimental groups. Group 1 (G1, n = 8) and group 2 (G2, n = 8) were dosed orally with 10 TgShSp1 sporulated oocysts at 90 days of gestation (dg). Animals from group 3 (G3, n = 3) were simultaneously mock dosed with phosphate-buffered solution (PBS). In G1, BKI-1748 was administered orally from day 7 p.i. (fever and increased serum IFNγ levels) onward, maintaining drug exposure for 20 days (10 doses at 15 mg/kg every 2 days). Treated animals (G1) exhibited significantly lower rectal temperatures (on days 8 and 9 p.i.), serum IFNγ levels (on day 10 p.i.), and specific IgG levels when compared with non-treated animals (G2). At delivery, significantly higher percentages of healthy lambs were found in infected/treated sheep in G1 (73.3%) and in uninfected sheep in G3 (80%) compared with infected/untreated sheep in G2 (31.3%). Concerning congenital transmission, parasite DNA was neither detected in placenta nor target tissues (brain and lungs) from the fetuses/lambs in G1(infected/treated) and G3 (uninfected). By contrast, parasite DNA was detected in all placentas and lambs from G2 (infected/untreated), except for one sheep that aborted on day 13 p.i.
Collapse
Affiliation(s)
- Roberto Sánchez-Sánchez
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, Madrid, Community of Madrid, Spain
| | - Ana Huertas-López
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, Madrid, Community of Madrid, Spain
- Animal Health Department, University of Murcia, Murcia, Region of Murcia, Spain
| | - Andrea Largo-de la Torre
- SALUVET-Innova S.L., Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, Madrid, Community of Madrid, Spain
| | - Ignacio Ferre
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, Madrid, Community of Madrid, Spain
| | - Filippo Maria Dini
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Emilia-Romagna, Italy
| | - Michela Re
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, Madrid, Community of Madrid, Spain
- Animal Medicine and Surgery Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, Madrid, Community of Madrid, Spain
| | - Javier Moreno-Gonzalo
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, Madrid, Community of Madrid, Spain
- Animal Medicine and Surgery Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, Madrid, Community of Madrid, Spain
| | - Ryan Choi
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Matthew A. Hulverson
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Kayode K. Ojo
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Samuel L. M. Arnold
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Berne, Berne, Switzerland
| | - Wesley C. Van Voorhis
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Luis Miguel Ortega-Mora
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, Madrid, Community of Madrid, Spain
| |
Collapse
|
3
|
O’Shaughnessy WJ, Dewangan PS, Paiz EA, Reese ML. Not your Mother's MAPKs: Apicomplexan MAPK function in daughter cell budding. PLoS Pathog 2022; 18:e1010849. [PMID: 36227859 PMCID: PMC9560070 DOI: 10.1371/journal.ppat.1010849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Reversible phosphorylation by protein kinases is one of the core mechanisms by which biological signals are propagated and processed. Mitogen-activated protein kinases, or MAPKs, are conserved throughout eukaryotes where they regulate cell cycle, development, and stress response. Here, we review advances in our understanding of the function and biochemistry of MAPK signaling in apicomplexan parasites. As expected for well-conserved signaling modules, MAPKs have been found to have multiple essential roles regulating both Toxoplasma tachyzoite replication and sexual differentiation in Plasmodium. However, apicomplexan MAPK signaling is notable for the lack of the canonical kinase cascade that normally regulates the networks, and therefore must be regulated by a distinct mechanism. We highlight what few regulatory relationships have been established to date, and discuss the challenges to the field in elucidating the complete MAPK signaling networks in these parasites.
Collapse
Affiliation(s)
- William J. O’Shaughnessy
- Department of Pharmacology, University of Texas, Southwestern Medical Center, Dallas, Texas, United States of America
| | - Pravin S. Dewangan
- Department of Pharmacology, University of Texas, Southwestern Medical Center, Dallas, Texas, United States of America
| | - E. Ariana Paiz
- Department of Pharmacology, University of Texas, Southwestern Medical Center, Dallas, Texas, United States of America
| | - Michael L. Reese
- Department of Pharmacology, University of Texas, Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Biochemistry, University of Texas, Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
4
|
Kaur P, Goyal N. Pathogenic role of mitogen activated protein kinases in protozoan parasites. Biochimie 2021; 193:78-89. [PMID: 34706251 DOI: 10.1016/j.biochi.2021.10.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/29/2021] [Accepted: 10/21/2021] [Indexed: 01/18/2023]
Abstract
Protozoan parasites with complex life cycles have high mortality rates affecting billions of human lives. Available anti-parasitic drugs are inadequate due to variable efficacy, toxicity, poor patient compliance and drug-resistance. Hence, there is an urgent need for the development of safer and better chemotherapeutics. Mitogen Activated Protein Kinases (MAPKs) have drawn much attention as potential drug targets. This review summarizes unique structural and functional features of MAP kinases and their possible role in pathogenesis of obligate intracellular protozoan parasites namely, Leishmania, Trypanosoma, Plasmodium and Toxoplasma. It also provides an overview of available knowledge concerning the target proteins of parasite MAPKs and the need to understand and unravel unknown interaction network(s) of MAPK(s).
Collapse
Affiliation(s)
- Pavneet Kaur
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Neena Goyal
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India.
| |
Collapse
|
5
|
Abstract
Mitogen-activated protein kinases (MAPKs) are a conserved family of protein kinases that regulate signal transduction, proliferation, and development throughout eukaryotes. The apicomplexan parasite Toxoplasma gondii expresses three MAPKs. Two of these, extracellular signal-regulated kinase 7 (ERK7) and MAPKL1, have been implicated in the regulation of conoid biogenesis and centrosome duplication, respectively. The third kinase, MAPK2, is specific to and conserved throughout the Alveolata, although its function is unknown. We used the auxin-inducible degron system to determine phenotypes associated with MAPK2 loss of function in Toxoplasma We observed that parasites lacking MAPK2 failed to duplicate their centrosomes and therefore did not initiate daughter cell budding, which ultimately led to parasite death. MAPK2-deficient parasites initiated but did not complete DNA replication and arrested prior to mitosis. Surprisingly, the parasites continued to grow and replicate their Golgi apparatus, mitochondria, and apicoplasts. We found that the failure in centrosome duplication is distinct from the phenotype caused by the depletion of MAPKL1. As we did not observe MAPK2 localization at the centrosome at any point in the cell cycle, our data suggest that MAPK2 regulates a process at a distal site that is required for the completion of centrosome duplication and the initiation of parasite mitosis.IMPORTANCE Toxoplasma gondii is a ubiquitous intracellular protozoan parasite that can cause severe and fatal disease in immunocompromised patients and the developing fetus. Rapid parasite replication is critical for establishing a productive infection. Here, we demonstrate that a Toxoplasma protein kinase called MAPK2 is conserved throughout the Alveolata and essential for parasite replication. We found that parasites lacking MAPK2 protein were defective in the initiation of daughter cell budding and were rendered inviable. Specifically, T. gondii MAPK2 (TgMAPK2) appears to be required for centrosome replication at the basal end of the nucleus, and its loss causes arrest early in parasite division. MAPK2 is unique to the Alveolata and not found in metazoa and likely is a critical component of an essential parasite-specific signaling network.
Collapse
|
6
|
Gubbels MJ, Keroack CD, Dangoudoubiyam S, Worliczek HL, Paul AS, Bauwens C, Elsworth B, Engelberg K, Howe DK, Coppens I, Duraisingh MT. Fussing About Fission: Defining Variety Among Mainstream and Exotic Apicomplexan Cell Division Modes. Front Cell Infect Microbiol 2020; 10:269. [PMID: 32582569 PMCID: PMC7289922 DOI: 10.3389/fcimb.2020.00269] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 05/06/2020] [Indexed: 12/17/2022] Open
Abstract
Cellular reproduction defines life, yet our textbook-level understanding of cell division is limited to a small number of model organisms centered around humans. The horizon on cell division variants is expanded here by advancing insights on the fascinating cell division modes found in the Apicomplexa, a key group of protozoan parasites. The Apicomplexa display remarkable variation in offspring number, whether karyokinesis follows each S/M-phase or not, and whether daughter cells bud in the cytoplasm or bud from the cortex. We find that the terminology used to describe the various manifestations of asexual apicomplexan cell division emphasizes either the number of offspring or site of budding, which are not directly comparable features and has led to confusion in the literature. Division modes have been primarily studied in two human pathogenic Apicomplexa, malaria-causing Plasmodium spp. and Toxoplasma gondii, a major cause of opportunistic infections. Plasmodium spp. divide asexually by schizogony, producing multiple daughters per division round through a cortical budding process, though at several life-cycle nuclear amplifications stages, are not followed by karyokinesis. T. gondii divides by endodyogeny producing two internally budding daughters per division round. Here we add to this diversity in replication mechanisms by considering the cattle parasite Babesia bigemina and the pig parasite Cystoisospora suis. B. bigemina produces two daughters per division round by a “binary fission” mechanism whereas C. suis produces daughters through both endodyogeny and multiple internal budding known as endopolygeny. In addition, we provide new data from the causative agent of equine protozoal myeloencephalitis (EPM), Sarcocystis neurona, which also undergoes endopolygeny but differs from C. suis by maintaining a single multiploid nucleus. Overall, we operationally define two principally different division modes: internal budding found in cyst-forming Coccidia (comprising endodyogeny and two forms of endopolygeny) and external budding found in the other parasites studied (comprising the two forms of schizogony, binary fission and multiple fission). Progressive insights into the principles defining the molecular and cellular requirements for internal vs. external budding, as well as variations encountered in sexual stages are discussed. The evolutionary pressures and mechanisms underlying apicomplexan cell division diversification carries relevance across Eukaryota.
Collapse
Affiliation(s)
- Marc-Jan Gubbels
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Caroline D Keroack
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, United States
| | - Sriveny Dangoudoubiyam
- Department of Veterinary Science, Gluck Equine Research Center, University of Kentucky, Lexington, KY, United States
| | - Hanna L Worliczek
- Department of Biology, Boston College, Chestnut Hill, MA, United States.,Institute of Parasitology, University of Veterinary Medicine, Vienna, Austria
| | - Aditya S Paul
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, United States
| | - Ciara Bauwens
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Brendan Elsworth
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, United States.,School of Biosciences, University of Melbourne, Melbourne, VIC, Australia
| | - Klemens Engelberg
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Daniel K Howe
- Department of Veterinary Science, Gluck Equine Research Center, University of Kentucky, Lexington, KY, United States
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Manoj T Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, United States
| |
Collapse
|
7
|
Liu S, Wu M, Hua Q, Lu D, Tian Y, Yu H, Cheng L, Chen Y, Cao J, Hu X, Tan F. Two old drugs, NVP-AEW541 and GSK-J4, repurposed against the Toxoplasma gondii RH strain. Parasit Vectors 2020; 13:242. [PMID: 32393321 PMCID: PMC7216583 DOI: 10.1186/s13071-020-04094-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/24/2020] [Indexed: 11/24/2022] Open
Abstract
Background Toxoplasma gondii is a zoonotic pathogen that causes toxoplasmosis and leads to serious public health problems in developing countries. However, current clinical therapeutic drugs have some disadvantages, such as serious side effects, a long course of treatment and the emergence of drug-resistant strains. The urgent need to identify novel anti-Toxoplasma drugs has initiated the effective strategy of repurposing well-characterized drugs. As a principled screening for the identification of effective compounds against Toxoplasma gondii, in the current study, a collection of 666 compounds were screened for their ability to significantly inhibit Toxoplasma growth. Methods The inhibition of parasite growth was determined using a luminescence-based β-galactosidase activity assay. Meanwhile, the effect of compounds on the viability of host cells was measured using CCK8. To assess the inhibition of the selected compounds on discrete steps of the T. gondii lytic cycle, the invasion, intracellular proliferation and egress abilities were evaluated. Finally, a murine infection model of toxoplasmosis was used to monitor the protective efficacy of drugs against acute infection of a highly virulent RH strain. Results A total of 68 compounds demonstrated more than 70% parasite growth inhibition. After excluding compounds that impaired host cell viability, we further characterized two compounds, NVP-AEW541 and GSK-J4 HCl, which had IC50 values for parasite growth of 1.17 μM and 2.37 μM, respectively. In addition, both compounds showed low toxicity to the host cell. Furthermore, we demonstrated that NVP-AEW541 inhibits tachyzoite invasion, while GSK-J4 HCl inhibits intracellular tachyzoite proliferation by halting cell cycle progression from G1 to S phase. These findings prompted us to analyse the efficacy of the two compounds in vivo by using established mouse models of acute toxoplasmosis. In addition to prolonging the survival time of mice acutely infected with T. gondii, both compounds had a remarkable ability to reduce the parasite burden of tissues. Conclusions Our findings suggest that both NVP-AEW541 and GSK-J4 could be potentially repurposed as candidate drugs against T. gondii infection.![]()
Collapse
Affiliation(s)
- Shuxian Liu
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Mimi Wu
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Qianqian Hua
- Clinical Laboratory, Dongyang People's Hospital, Jinhua, 322100, Zhejiang, People's Republic of China
| | - Daiqiang Lu
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yuan Tian
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Helin Yu
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Linyan Cheng
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yinqi Chen
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jiaxin Cao
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xin Hu
- School of Medical Laboratory Science and School of Life Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Feng Tan
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
8
|
Hammarton TC. Who Needs a Contractile Actomyosin Ring? The Plethora of Alternative Ways to Divide a Protozoan Parasite. Front Cell Infect Microbiol 2019; 9:397. [PMID: 31824870 PMCID: PMC6881465 DOI: 10.3389/fcimb.2019.00397] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/06/2019] [Indexed: 01/21/2023] Open
Abstract
Cytokinesis, or the division of the cytoplasm, following the end of mitosis or meiosis, is accomplished in animal cells, fungi, and amoebae, by the constriction of an actomyosin contractile ring, comprising filamentous actin, myosin II, and associated proteins. However, despite this being the best-studied mode of cytokinesis, it is restricted to the Opisthokonta and Amoebozoa, since members of other evolutionary supergroups lack myosin II and must, therefore, employ different mechanisms. In particular, parasitic protozoa, many of which cause significant morbidity and mortality in humans and animals as well as considerable economic losses, employ a wide diversity of mechanisms to divide, few, if any, of which involve myosin II. In some cases, cell division is not only myosin II-independent, but actin-independent too. Mechanisms employed range from primitive mechanical cell rupture (cytofission), to motility- and/or microtubule remodeling-dependent mechanisms, to budding involving the constriction of divergent contractile rings, to hijacking host cell division machinery, with some species able to utilize multiple mechanisms. Here, I review current knowledge of cytokinesis mechanisms and their molecular control in mammalian-infective parasitic protozoa from the Excavata, Alveolata, and Amoebozoa supergroups, highlighting their often-underappreciated diversity and complexity. Billions of people and animals across the world are at risk from these pathogens, for which vaccines and/or optimal treatments are often not available. Exploiting the divergent cell division machinery in these parasites may provide new avenues for the treatment of protozoal disease.
Collapse
Affiliation(s)
- Tansy C Hammarton
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
9
|
Montazeri M, Mehrzadi S, Sharif M, Sarvi S, Tanzifi A, Aghayan SA, Daryani A. Drug Resistance in Toxoplasma gondii. Front Microbiol 2018; 9:2587. [PMID: 30420849 PMCID: PMC6215853 DOI: 10.3389/fmicb.2018.02587] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 10/10/2018] [Indexed: 12/12/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is a global protozoan parasite infecting up to one-third of the world population. Pyrimethamine (PYR) and sulfadiazine (SDZ) are the most widely used drugs for treatment of toxoplasmosis; however, several failure cases have been recorded as well; suggesting the existence of drug resistant strains. This review aims to give a systematic and comprehensive understanding of drug resistance in T. gondii including mechanisms of resistance and sites of drug action in parasite. Analogous amino acid substitutions in the Toxoplasma enzyme were identified to confer PYR resistance. Moreover, resistance to clindamycin, spiramycin, and azithromycin is encoded in the rRNA genes of T. gondii. However, T. gondii SDZ resistance mechanism has not been proved yet. Recently there has been a slight increase in SDZ resistance. That is why the majority of studies were carried out using SDZ. Six strains resistant to SDZ were found in clinical cases between 2013 and 2017 which among Brazilian T. gondii isolates, TgCTBr11, Ck3, and Pg1 were identified in human toxoplasmosis, as well as in livestock intended for human consumption. In conclusion, recent experimental studies in clinical cases have clearly shown that drug resistance in Toxoplasma is ongoing. Thus, establishing a more effective therapeutic scheme in the treatment of toxoplasmosis is critically needed. The emergence of T. gondii strains resistant to current drugs, reviewed here, represents a concern not only for treatment failure but also for increased clinical severity in immunocompromised patients. To improve the therapeutic outcome in patients, a greater understanding of the exact mechanisms of drug resistance in T. gondii should be developed. Thus, monitoring the presence of resistant parasites, in food products, would seem a prudent public health program.
Collapse
Affiliation(s)
- Mahbobeh Montazeri
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Sharif
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Parasitology, School of Medicine, Sari Branch, Islamic Azad University, Sari, Iran
| | - Shahabeddin Sarvi
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Parasitology, Sari Medical School, Mazandaran University of Medical Sciences, Sari, Iran
| | - Asal Tanzifi
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sargis A Aghayan
- Laboratory of Zoology, Research Institute of Biology, Yerevan State University, Yerevan, Armenia
| | - Ahmad Daryani
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Parasitology, Sari Medical School, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
10
|
Tu V, Yakubu R, Weiss LM. Observations on bradyzoite biology. Microbes Infect 2018; 20:466-476. [PMID: 29287987 PMCID: PMC6019562 DOI: 10.1016/j.micinf.2017.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/13/2017] [Accepted: 12/15/2017] [Indexed: 02/06/2023]
Abstract
Tachyzoites of the Apicomplexan Toxoplasma gondii cause acute infection, disseminate widely in their host, and eventually differentiate into a latent encysted form called bradyzoites that are found within tissue cysts. During latent infection, whenever transformation to tachyzoites occurs, any tachyzoites that develop are removed by the immune system. In contrast, cysts containing bradyzoites are sequestered from the immune system. In the absence of an effective immune response released organisms that differentiate into tachyzoites cause acute infection. Tissue cysts, therefore, serve as a reservoir for the reactivation of toxoplasmosis when the host becomes immunocompromised by conditions such as HIV infection, organ transplantation, or due to the impaired immune response that occurs when pathogens are acquired in utero. While tachyzoites and bradyzoites are well defined morphologically, there is no clear consensus on how interconversion occurs or what exact signal(s) mediate this transformation. Advances in research methods have facilitated studies on T. gondii bradyzoites providing important new insights into the biology of latent infection.
Collapse
Affiliation(s)
- Vincent Tu
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rama Yakubu
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Louis M Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
11
|
Toxoplasma Calcium-Dependent Protein Kinase 1 Inhibitors: Probing Activity and Resistance Using Cellular Thermal Shift Assays. Antimicrob Agents Chemother 2018; 62:AAC.00051-18. [PMID: 29555627 DOI: 10.1128/aac.00051-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 03/12/2018] [Indexed: 11/20/2022] Open
Abstract
In Toxoplasma gondii, calcium-dependent protein kinase 1 (CDPK1) is an essential protein kinase required for invasion of host cells. We have developed several hundred CDPK1 inhibitors, many of which block invasion. Inhibitors with similar 50% inhibitory concentrations (IC50s) were tested in thermal shift assays for their ability to stabilize CDPK1 in cell lysates, in intact cells, or in purified form. Compounds that inhibited parasite growth stabilized CDPK1 in all assays. In contrast, two compounds that showed poor growth inhibition stabilized CDPK1 in lysates but not in cells. Thus, cellular exclusion could explain exceptions in the correlation between the action on the target and cellular activity. We used thermal shift assays to examine CDPK1 in two clones that were independently selected by growth in the CDPK1 inhibitor RM-1-132 and that had increased 50% effective concentrations (EC50s) for the compound. The A and C clones had distinct point mutations in the CDPK1 kinase domain, H201Q and L96P, respectively, residues that lie near one another in the inactive isoform. Purified mutant proteins showed RM-1-132 IC50s and thermal shifts similar to those shown by wild-type CDPK1. Reduced inhibitor stabilization (and a presumed reduced interaction) was observed only in cellular thermal shift assays. This highlights the utility of cellular thermal shift assays in demonstrating that resistance involves reduced on-target engagement (even if biochemical assays suggest otherwise). Indeed, similar EC50s were observed upon overexpression of the mutant proteins, as in the corresponding drug-selected parasites, although high levels of CDPK1(H201Q) only modestly increased resistance compared to that achieved with high levels of wild-type enzyme.
Collapse
|
12
|
In vitro efficacy of bumped kinase inhibitors against Besnoitia besnoiti tachyzoites. Int J Parasitol 2017; 47:811-821. [PMID: 28899692 DOI: 10.1016/j.ijpara.2017.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/25/2017] [Accepted: 08/29/2017] [Indexed: 12/30/2022]
Abstract
Besnoitia besnoiti is an apicomplexan parasite responsible for bovine besnoitiosis, a chronic and debilitating disease that causes systemic and skin manifestations and sterility in bulls. Neither treatments nor vaccines are currently available. In the search for therapeutic candidates, calcium-dependent protein kinases have arisen as promising drug targets in other apicomplexans (e.g. Neospora caninum, Toxoplasma gondii, Plasmodium spp. and Eimeria spp.) and are effectively targeted by bumped kinase inhibitors. In this study, we identified and cloned the gene coding for BbCDPK1. The impact of a library of nine bumped kinase inhibitor analogues on the activity of recombinant BbCDPK1 was assessed by luciferase assay. Afterwards, those were further screened for efficacy against Besnoitiabesnoiti tachyzoites grown in Marc-145 cells. Primary tests at 5µM revealed that eight compounds exhibited more than 90% inhibition of invasion and proliferation. The compounds BKI 1294, 1517, 1553 and 1571 were further characterised, and EC99 (1294: 2.38µM; 1517: 2.20µM; 1553: 3.34µM; 1571: 2.78µM) were determined by quantitative real-time polymerase chain reaction in 3-day proliferation assays. Exposure of infected cultures with EC99 concentrations of these drugs for up to 48h was not parasiticidal. The lack of parasiticidal action was confirmed by transmission electron microscopy, which showed that bumped kinase inhibitor treatment interfered with cell cycle regulation and non-disjunction of tachyzoites, resulting in the formation of large multi-nucleated complexes which co-existed with viable parasites within the parasitophorous vacuole. However, it is possible that, in the face of an active immune response, parasite clearance may occur. In summary, bumped kinase inhibitors may be effective drug candidates to control Besnoitiabesnoiti infection. Further in vivo experiments should be planned, as attainment and maintenance of therapeutic blood plasma levels in calves, without toxicity, has been demonstrated for BKIs 1294, 1517 and 1553.
Collapse
|
13
|
Alday PH, Doggett JS. Drugs in development for toxoplasmosis: advances, challenges, and current status. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:273-293. [PMID: 28182168 PMCID: PMC5279849 DOI: 10.2147/dddt.s60973] [Citation(s) in RCA: 230] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Toxoplasma gondii causes fatal and debilitating brain and eye diseases. Medicines that are currently used to treat toxoplasmosis commonly have toxic side effects and require prolonged courses that range from weeks to more than a year. The need for long treatment durations and the risk of relapsing disease are in part due to the lack of efficacy against T. gondii tissue cysts. The challenges for developing a more effective treatment for toxoplasmosis include decreasing toxicity, achieving therapeutic concentrations in the brain and eye, shortening duration, eliminating tissue cysts from the host, safety in pregnancy, and creating a formulation that is inexpensive and practical for use in resource-poor areas of the world. Over the last decade, significant progress has been made in identifying and developing new compounds for the treatment of toxoplasmosis. Unlike clinically used medicines that were repurposed for toxoplasmosis, these compounds have been optimized for efficacy against toxoplasmosis during preclinical development. Medicines with enhanced efficacy as well as features that address the unique aspects of toxoplasmosis have the potential to greatly improve toxoplasmosis therapy. This review discusses the facets of toxoplasmosis that are pertinent to drug design and the advances, challenges, and current status of preclinical drug research for toxoplasmosis.
Collapse
Affiliation(s)
- P Holland Alday
- Division of Infectious Diseases, Oregon Health & Science University
| | - Joseph Stone Doggett
- Division of Infectious Diseases, Oregon Health & Science University; Portland Veterans Affairs Medical Center, Portland, OR, USA
| |
Collapse
|
14
|
Van Voorhis WC, Doggett JS, Parsons M, Hulverson MA, Choi R, Arnold SLM, Riggs MW, Hemphill A, Howe DK, Mealey RH, Lau AOT, Merritt EA, Maly DJ, Fan E, Ojo KK. Extended-spectrum antiprotozoal bumped kinase inhibitors: A review. Exp Parasitol 2017; 180:71-83. [PMID: 28065755 DOI: 10.1016/j.exppara.2017.01.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 01/02/2017] [Accepted: 01/04/2017] [Indexed: 01/08/2023]
Abstract
Many life-cycle processes in parasites are regulated by protein phosphorylation. Hence, disruption of essential protein kinase function has been explored for therapy of parasitic diseases. However, the difficulty of inhibiting parasite protein kinases to the exclusion of host orthologues poses a practical challenge. A possible path around this difficulty is the use of bumped kinase inhibitors for targeting calcium-dependent protein kinases that contain atypically small gatekeeper residues and are crucial for pathogenic apicomplexan parasites' survival and proliferation. In this article, we review efficacy against the kinase target, parasite growth in vitro, and in animal infection models, as well as the relevant pharmacokinetic and safety parameters of bumped kinase inhibitors.
Collapse
Affiliation(s)
- Wesley C Van Voorhis
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious Diseases (CERID), University of Washington, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Seattle, WA 98195, USA.
| | | | - Marilyn Parsons
- Department of Global Health, University of Washington, Seattle, WA 98195, USA; Center for Infectious Disease Research, Seattle, WA 98109, USA
| | - Matthew A Hulverson
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious Diseases (CERID), University of Washington, Seattle, WA 98109, USA
| | - Ryan Choi
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious Diseases (CERID), University of Washington, Seattle, WA 98109, USA
| | - Samuel L M Arnold
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious Diseases (CERID), University of Washington, Seattle, WA 98109, USA
| | - Michael W Riggs
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Berne, Switzerland
| | - Daniel K Howe
- Department of Veterinary Science, University of Kentucky, Lexington, KY 40546, USA
| | - Robert H Mealey
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-7040, USA
| | - Audrey O T Lau
- The National Institutes of Health, NIAID, DEA, 5601 Fishers Lane, Rockville, MD 20892, USA
| | - Ethan A Merritt
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Dustin J Maly
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Erkang Fan
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Kayode K Ojo
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious Diseases (CERID), University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
15
|
Kato K, Sugi T, Takemae H, Takano R, Gong H, Ishiwa A, Horimoto T, Akashi H. Characterization of a Toxoplasma gondii calcium calmodulin-dependent protein kinase homolog. Parasit Vectors 2016; 9:405. [PMID: 27444499 PMCID: PMC4957278 DOI: 10.1186/s13071-016-1676-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 06/30/2016] [Indexed: 12/04/2022] Open
Abstract
Background Toxoplasma gondii is an obligate intracellular parasite of the phylum Apicomplexa and a major pathogen of animals and immunocompromised humans, in whom it causes encephalitis. Understanding the mechanism of tachyzoite invasion is important for the discovery of new drug targets and may serve as a model for the study of other apicomplexan parasites. We previously showed that Plasmodium falciparum expresses a homolog of human calcium calmodulin-dependent protein kinase (CaMK) that is important for host cell invasion. In this study, to identify novel targets for the treatment of Toxoplasma gondii infection (another apicomplexan parasite), we sought to identify a CaMK-like protein in the T. gondii genome and to characterize its role in the life-cycle of this parasite. Methods An in vitro kinase assay was performed to assess the phosphorylation activities of a novel CaMK-like protein in T. gondii by using purified proteins with various concentrations of calcium, calmodulin antagonists, or T. gondii glideosome proteins. Indirect immunofluorescence microscopy was performed to detect the localization of this protein kinase by using the antibodies against this protein and organellar maker proteins of T. gondii. Results We identified a novel CaMK homolog in T. gondii, T. gondii CaMK-related kinase (TgCaMKrk), which exhibits calmodulin-independent autophosphorylation and substrate phosphorylation activity. However, calmodulin antagonists had no effect on its kinase activity. In T. gondii-infected cells, TgCaMKrk localized to the apical ends of extracellular and intracellular tachyzoites. TgCaMKrk phosphorylated TgGAP45 for phosphorylation in vitro. Conclusions Our data improve our understanding of T. gondii motility and infection, the interaction between parasite protein kinases and glideosomes, and drug targets for protozoan diseases.
Collapse
Affiliation(s)
- Kentaro Kato
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan. .,Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan.
| | - Tatsuki Sugi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan.,Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Hitoshi Takemae
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan.,Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Ryo Takano
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Haiyan Gong
- Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Akiko Ishiwa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan.,Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Taisuke Horimoto
- Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Hiroomi Akashi
- Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| |
Collapse
|
16
|
Toxoplasma gondii Cyclic AMP-Dependent Protein Kinase Subunit 3 Is Involved in the Switch from Tachyzoite to Bradyzoite Development. mBio 2016; 7:mBio.00755-16. [PMID: 27247232 PMCID: PMC4895117 DOI: 10.1128/mbio.00755-16] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Toxoplasma gondii is an obligate intracellular apicomplexan parasite that infects warm-blooded vertebrates, including humans. Asexual reproduction in T. gondii allows it to switch between the rapidly replicating tachyzoite and quiescent bradyzoite life cycle stages. A transient cyclic AMP (cAMP) pulse promotes bradyzoite differentiation, whereas a prolonged elevation of cAMP inhibits this process. We investigated the mechanism(s) by which differential modulation of cAMP exerts a bidirectional effect on parasite differentiation. There are three protein kinase A (PKA) catalytic subunits (TgPKAc1 to -3) expressed in T. gondii. Unlike TgPKAc1 and TgPKAc2, which are conserved in the phylum Apicomplexa, TgPKAc3 appears evolutionarily divergent and specific to coccidian parasites. TgPKAc1 and TgPKAc2 are distributed in the cytomembranes, whereas TgPKAc3 resides in the cytosol. TgPKAc3 was genetically ablated in a type II cyst-forming strain of T. gondii (PruΔku80Δhxgprt) and in a type I strain (RHΔku80Δhxgprt), which typically does not form cysts. The Δpkac3 mutant exhibited slower growth than the parental and complemented strains, which correlated with a higher basal rate of tachyzoite-to-bradyzoite differentiation. 3-Isobutyl-1-methylxanthine (IBMX) treatment, which elevates cAMP levels, maintained wild-type parasites as tachyzoites under bradyzoite induction culture conditions (pH 8.2/low CO2), whereas the Δpkac3 mutant failed to respond to the treatment. This suggests that TgPKAc3 is the factor responsible for the cAMP-dependent tachyzoite maintenance. In addition, the Δpkac3 mutant had a defect in the production of brain cysts in vivo, suggesting that a substrate of TgPKAc3 is probably involved in the persistence of this parasite in the intermediate host animals. Toxoplasma gondii is one of the most prevalent eukaryotic parasites in mammals, including humans. Parasites can switch from rapidly replicating tachyzoites responsible for acute infection to slowly replicating bradyzoites that persist as a latent infection. Previous studies have demonstrated that T. gondii cAMP signaling can induce or suppress bradyzoite differentiation, depending on the strength and duration of cAMP signal. Here, we report that TgPKAc3 is responsible for cAMP-dependent tachyzoite maintenance while suppressing differentiation into bradyzoites, revealing one mechanism underlying how this parasite transduces cAMP signals during differentiation.
Collapse
|
17
|
Engelberg K, Ivey FD, Lin A, Kono M, Lorestani A, Faugno-Fusci D, Gilberger TW, White M, Gubbels MJ. A MORN1-associated HAD phosphatase in the basal complex is essential for Toxoplasma gondii daughter budding. Cell Microbiol 2016; 18:1153-71. [PMID: 26840427 DOI: 10.1111/cmi.12574] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 12/24/2015] [Accepted: 01/19/2016] [Indexed: 12/30/2022]
Abstract
Apicomplexan parasites replicate by several budding mechanisms with two well-characterized examples being Toxoplasma endodyogeny and Plasmodium schizogony. Completion of budding requires the tapering of the nascent daughter buds toward the basal end, driven by contraction of the basal complex. This contraction is not executed by any of the known cell division associated contractile mechanisms and in order to reveal new components of the unusual basal complex we performed a yeast two-hybrid screen with its major scaffolding protein, TgMORN1. Here we report on a conserved protein with a haloacid dehalogenase (HAD) phosphatase domain, hereafter named HAD2a, identified by yeast two-hybrid. HAD2a has demonstrated enzyme-activity in vitro, localizes to the nascent daughter buds, and co-localizes with MORN1 to the basal complex during its contraction. Conditional knockout of HAD2a in Toxoplasma interferes with basal complex assembly, which leads to incomplete cytokinesis and conjoined daughters that ultimately results in disrupted proliferation. In Plasmodium, we further confirmed localization of the HAD2a ortholog to the basal complex toward the end of schizogony. In conclusion, our work highlights an essential role for this HAD phosphatase across apicomplexan budding and suggests a regulatory mechanism of differential phosphorylation on the structure and/or contractile function of the basal complex.
Collapse
Affiliation(s)
- Klemens Engelberg
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
| | - F Douglas Ivey
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
| | - Angela Lin
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
| | - Maya Kono
- Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany
| | | | - Dave Faugno-Fusci
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
| | - Tim-Wolf Gilberger
- Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany.,M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.,Center for Structural Systems Biology, Hamburg, Germany
| | - Michael White
- Departments of Molecular Medicine & Global Health, Florida Center for Drug Discovery and Innovation, Colleges of Medicine and Public Health, University of South Florida, Tampa, FL, 33612, USA
| | - Marc-Jan Gubbels
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
| |
Collapse
|
18
|
In Vitro and In Vivo Effects of the Bumped Kinase Inhibitor 1294 in the Related Cyst-Forming Apicomplexans Toxoplasma gondii and Neospora caninum. Antimicrob Agents Chemother 2015; 59:6361-74. [PMID: 26248379 DOI: 10.1128/aac.01236-15] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 07/22/2015] [Indexed: 12/11/2022] Open
Abstract
We report on the in vitro effects of the bumped kinase inhibitor 1294 (BKI-1294) in cultures of virulent Neospora caninum isolates Nc-Liverpool (Nc-Liv) and Nc-Spain7 and in two strains of Toxoplasma gondii (RH and ME49), all grown in human foreskin fibroblasts. In these parasites, BKI-1294 acted with 50% inhibitory concentrations (IC50s) ranging from 20 nM (T. gondii RH) to 360 nM (N. caninum Nc-Liv), and exposure of intracellular stages to 1294 led to the nondisjunction of newly formed tachyzoites, resulting in the formation of multinucleated complexes similar to complexes previously observed in BKI-1294-treated N. caninum beta-galactosidase-expressing parasites. However, such complexes were not seen in a transgenic T. gondii strain that expressed CDPK1 harboring a mutation (G to M) in the gatekeeper residue. In T. gondii ME49 and N. caninum Nc-Liv, exposure of cultures to BKI-1294 resulted in the elevated expression of mRNA coding for the bradyzoite marker BAG1. Unlike in bradyzoites, SAG1 expression was not repressed. Immunofluorescence also showed that these multinucleated complexes expressed SAG1 and BAG1 and the monoclonal antibody CC2, which binds to a yet unidentified bradyzoite antigen, also exhibited increased labeling. In a pregnant mouse model, BKI-1294 efficiently inhibited vertical transmission in BALB/c mice experimentally infected with one of the two virulent isolates Nc-Liv or Nc-Spain7, demonstrating proof of concept that this compound protected offspring from vertical transmission and disease. The observed deregulated antigen expression effect may enhance the immune response during BKI-1294 therapy and will be the subject of future studies.
Collapse
|