1
|
Minutti AF, Sasse JP, Dos Santos Silva AC, Martins TA, Martinez V, de Souza Lima Nino B, de Souza Rodrigues F, de Barros LD, Garcia JL. Evaluation of a DNA vector plasmid encoding a partial rop18 gene from toxoplasma gondii in domestic cats as a vaccine candidate. Vaccine 2025; 54:126965. [PMID: 40054140 DOI: 10.1016/j.vaccine.2025.126965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 05/13/2025]
Abstract
The present study aimed to evaluate a DNA vector plasmid encoding a partial rop18 gene from Toxoplasma gondii in domestic cats as a potential vaccine candidate. Four domestic cats (Felis catus) were used, of which two animals received 25 μg of pcDNA 3.1 + rop18, and two received 25 μg of pcDNA 3.1. All animals received intramuscular immunizations with four doses every three weeks along with 1.5 % levamisole. Thirty days after the last immunization, the animals were infected with 300 tissue cysts from ToxoDB #182 strain, a non-archetypal genotype isolated from a wild cat. Fecal examinations were performed for oocyst shedding. Enzyme-linked immunosorbent assay and western blotting analyses with recombinant ROP18 were performed to assess the humoral immune response. Animals that received plasmid containing the partial T. gondii rop18 gene produced specific IgG antibodies and shed 53.3 % fewer oocysts than controls. The two groups of animals showed no statistically significant differences (p > 0.05) in oocyst shedding; however, they showed significant differences in the detection of anti-Toxoplasma antibodies (p < 0.05). In conclusion, the T. gondii rop18 gene is a potential vaccine candidate against oocyst shedding in cats.
Collapse
Affiliation(s)
- Ana Flávia Minutti
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - João Pedro Sasse
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - Ana Clécia Dos Santos Silva
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - Thais Agostinho Martins
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - Valentina Martinez
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - Beatriz de Souza Lima Nino
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - Fernando de Souza Rodrigues
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - Luiz Daniel de Barros
- Laboratory of Veterinary Parasitology and Parasitic Diseases, Universidade Federal de Lavras - UFLA, Lavras, MG, Brazil
| | - João Luis Garcia
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil.
| |
Collapse
|
2
|
Lykins J, Moschitto MJ, Zhou Y, Filippova EV, Le HV, Tomita T, Fox BA, Bzik DJ, Su C, Rajagopala SV, Flores K, Spano F, Woods S, Roberts CW, Hua C, El Bissati K, Wheeler KM, Dovgin S, Muench SP, McPhillie M, Fishwick CW, Anderson WF, Lee PJ, Hickman M, Weiss LM, Dubey JP, Lorenzi HA, Silverman RB, McLeod RL. From TgO/GABA-AT, GABA, and T-263 Mutant to Conception of Toxoplasma. iScience 2024; 27:108477. [PMID: 38205261 PMCID: PMC10776954 DOI: 10.1016/j.isci.2023.108477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 04/28/2023] [Accepted: 11/13/2023] [Indexed: 01/12/2024] Open
Abstract
Toxoplasma gondii causes morbidity, mortality, and disseminates widely via cat sexual stages. Here, we find T. gondii ornithine aminotransferase (OAT) is conserved across phyla. We solve TgO/GABA-AT structures with bound inactivators at 1.55 Å and identify an inactivator selective for TgO/GABA-AT over human OAT and GABA-AT. However, abrogating TgO/GABA-AT genetically does not diminish replication, virulence, cyst-formation, or eliminate cat's oocyst shedding. Increased sporozoite/merozoite TgO/GABA-AT expression led to our study of a mutagenized clone with oocyst formation blocked, arresting after forming male and female gametes, with "Rosetta stone"-like mutations in genes expressed in merozoites. Mutations are similar to those in organisms from plants to mammals, causing defects in conception and zygote formation, affecting merozoite capacitation, pH/ionicity/sodium-GABA concentrations, drawing attention to cyclic AMP/PKA, and genes enhancing energy or substrate formation in TgO/GABA-AT-related-pathways. These candidates potentially influence merozoite's capacity to make gametes that fuse to become zygotes, thereby contaminating environments and causing disease.
Collapse
Affiliation(s)
- Joseph Lykins
- Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Matthew J. Moschitto
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, and Center for Developmental Therapeutics, Northwestern University, Evanston, IL 60208-3113, USA
| | - Ying Zhou
- Department of Ophthalmology and Visual Sciences, The University of Chicago, Chicago, IL 60637, USA
| | - Ekaterina V. Filippova
- Center for Structural Genomics of Infectious Diseases and the Department of Biochemistry and Molecular Genetics, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hoang V. Le
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, and Center for Developmental Therapeutics, Northwestern University, Evanston, IL 60208-3113, USA
| | - Tadakimi Tomita
- Division of Parasitology, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Barbara A. Fox
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - David J. Bzik
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Chunlei Su
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | - Seesandra V. Rajagopala
- Department of Infectious Diseases, The J. Craig Venter Institute, 9704 Medical Center Drive, Rockville, MD 20850, USA
| | - Kristin Flores
- Center for Structural Genomics of Infectious Diseases and the Department of Biochemistry and Molecular Genetics, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Furio Spano
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Stuart Woods
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow Scotland, UK
| | - Craig W. Roberts
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow Scotland, UK
| | - Cong Hua
- Department of Ophthalmology and Visual Sciences, The University of Chicago, Chicago, IL 60637, USA
| | - Kamal El Bissati
- Department of Ophthalmology and Visual Sciences, The University of Chicago, Chicago, IL 60637, USA
| | - Kelsey M. Wheeler
- Department of Ophthalmology and Visual Sciences, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah Dovgin
- Department of Ophthalmology and Visual Sciences, The University of Chicago, Chicago, IL 60637, USA
| | - Stephen P. Muench
- School of Biomedical Sciences and Astbury Centre for Structural Molecular Biology, The University of Leeds, Leeds, West York LS2 9JT, UK
| | - Martin McPhillie
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Colin W.G. Fishwick
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Wayne F. Anderson
- Center for Structural Genomics of Infectious Diseases and the Department of Biochemistry and Molecular Genetics, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - Patricia J. Lee
- Division of Experimental Therapeutics, Military Malaria Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Mark Hickman
- Division of Experimental Therapeutics, Military Malaria Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Louis M. Weiss
- Division of Parasitology, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jitender P. Dubey
- Animal Parasitic Diseases Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA
| | - Hernan A. Lorenzi
- Department of Infectious Diseases, The J. Craig Venter Institute, 9704 Medical Center Drive, Rockville, MD 20850, USA
| | - Richard B. Silverman
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, and Center for Developmental Therapeutics, Northwestern University, Evanston, IL 60208-3113, USA
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - Rima L. McLeod
- Department of Ophthalmology and Visual Sciences, The University of Chicago, Chicago, IL 60637, USA
- Department of Pediatrics (Infectious Diseases), Institute of Genomics, Genetics, and Systems Biology, Global Health Center, Toxoplasmosis Center, CHeSS, The College, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
3
|
Zhai B, Xie SC, Zhang J, He JJ, Zhu XQ. Dynamic RNA profiles in the small intestinal epithelia of cats after Toxoplasma gondii infection. Infect Dis Poverty 2023; 12:68. [PMID: 37491273 PMCID: PMC10367386 DOI: 10.1186/s40249-023-01121-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/14/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Felids are the only definitive hosts of Toxoplasma gondii. However, the biological features of the feline small intestine following T. gondii infection are poorly understood. We investigated the changes in the expression of RNAs (including mRNAs, long non-coding RNAs and circular RNAs) in the small intestinal epithelia of cats following T. gondii infection to improve our understanding of the life cycle of T. gondii and cat responses to T. gondii infection. METHODS Fifteen cats were randomly assigned to five groups, and the infection groups were inoculated with 600 tissue cysts of the T. gondii Pru strain by gavage. The small intestinal epithelia of cats were collected at 6, 10, 14, and 30 days post infection (DPI). Using high-throughput RNA sequencing (RNA-seq), we investigated the changes in RNA expression. The expression levels of differentially expressed (DE) genes and non-coding RNAs (ncRNAs) identified by RNA-seq were validated by quantitative reverse transcription PCR (qRT-PCR). Differential expression was determined using the DESeq R package. RESULTS In total, 207 annotated lncRNAs, 20,552 novel lncRNAs, 3342 novel circRNAs and 19,409 mRNAs were identified. Among these, 70 to 344 DE mRNAs, lncRNAs and circRNAs were detected, and the post-cleavage binding sites between 725 ncRNAs and 2082 miRNAs were predicted. Using the co-location method, we predicted that a total of 235 lncRNAs target 1044 protein-coding genes, while the results of co-expression analysis revealed that 174 lncRNAs target 2097 mRNAs. Pathway enrichment analyses of the genes targeted by ncRNAs suggested that most ncRNAs were significantly enriched in immune or diseases-related pathways. NcRNA regulatory networks revealed that a single ncRNA could be directly or indirectly regulated by multiple genes or ncRNAs that could influence the immune response of cats. Co-expression analysis showed that 242 circRNAs, mainly involved in immune responses, were significantly associated with T. gondii infection. In contrast, 1352 protein coding RNAs, mainly involved in nucleic acid process/repair pathways or oocyte development pathways, were negatively associated with T. gondii infection. CONCLUSIONS This study is the first to reveal the expression profiles of circRNAs, lncRNAs and mRNAs in the cat small intestine following T. gondii infection and will facilitate the elucidation of the role of ncRNAs in the pathogenesis of T. gondii infection in its definitive host, thereby facilitating the development of novel intervention strategies against T. gondii infection in humans and animals.
Collapse
Affiliation(s)
- Bintao Zhai
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, Gansu, People's Republic of China
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu, People's Republic of China
| | - Shi-Chen Xie
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, People's Republic of China
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, Hunan, People's Republic of China
| | - Jiyu Zhang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, Gansu, People's Republic of China
| | - Jun-Jun He
- Key Laboratory of Veterinary Public Health of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, People's Republic of China.
| | - Xing-Quan Zhu
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, People's Republic of China.
- Key Laboratory of Veterinary Public Health of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, People's Republic of China.
| |
Collapse
|
4
|
Srivastava S, Holmes MJ, White MW, Sullivan WJ. Toxoplasma gondii AP2XII-2 Contributes to Transcriptional Repression for Sexual Commitment. mSphere 2023; 8:e0060622. [PMID: 36786611 PMCID: PMC10117075 DOI: 10.1128/msphere.00606-22] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/15/2023] [Indexed: 02/15/2023] Open
Abstract
Toxoplasma gondii is a widespread protozoan parasite that has a significant impact on human and veterinary health. The parasite undergoes a complex life cycle involving multiple hosts and developmental stages. How Toxoplasma transitions between life cycle stages is poorly understood yet central to controlling transmission. Of particular neglect are the factors that contribute to its sexual development, which takes place exclusively in feline intestines. While epigenetic repressors have been shown to play an important role in silencing the spurious gene expression of sexually committed parasites, the specific factors that recruit this generalized machinery to the appropriate genes remain largely unexplored. Here, we establish that a member of the AP2 transcription factor family, AP2XII-2, is targeted to genomic loci associated with sexually committed parasites along with epigenetic regulators of transcriptional silencing, HDAC3 and MORC. Despite its widespread association with gene promoters, AP2XII-2 is required for the silencing of relatively few genes. Using the CUT&Tag (cleavage under targets and tagmentation) methodology, we identify two major genes associated with sexual development downstream of AP2XII-2 control, AP2X-10 and the amino acid hydroxylase AAH1. Our findings show that AP2XII-2 is a key contributor to the gene regulatory pathways modulating Toxoplasma sexual development. IMPORTANCE Toxoplasma gondii is a parasite that undergoes its sexual stage exclusively in feline intestines, making cats a major source of transmission. A better understanding of the proteins controlling the parasite's life cycle stage transitions is needed for the development of new therapies aimed at treating toxoplasmosis and the transmission of the infection. Genes that regulate the sexual stages need to be turned on and off at the appropriate times, activities that are mediated by specific transcription factors that recruit general machinery to silence or activate gene expression. In this study, we identify a transcription factor called AP2XII-2 as being important for the repression of a subset of sexual stage genes, including a sexual stage-specific AP2 factor (AP2X-10) and a protein (AAH1) required to construct the infectious oocysts expelled from infected cats.
Collapse
Affiliation(s)
- Sandeep Srivastava
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael J. Holmes
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael W. White
- Department of Global Health, University of South Florida, Tampa, Florida, USA
| | - William J. Sullivan
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
5
|
Late Embryogenesis Abundant Proteins Contribute to the Resistance of Toxoplasma gondii Oocysts against Environmental Stresses. mBio 2023; 14:e0286822. [PMID: 36809045 PMCID: PMC10128015 DOI: 10.1128/mbio.02868-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Toxoplasma gondii oocysts, which are shed in large quantities in the feces from infected felines, are very stable in the environment, resistant to most inactivation procedures, and highly infectious. The oocyst wall provides an important physical barrier for sporozoites contained inside oocysts, protecting them from many chemical and physical stressors, including most inactivation procedures. Furthermore, sporozoites can withstand large temperature changes, even freeze-thawing, as well as desiccation, high salinity, and other environmental insults; however, the genetic basis for this environmental resistance is unknown. Here, we show that a cluster of four genes encoding Late Embryogenesis Abundant (LEA)-related proteins are required to provide Toxoplasma sporozoites resistance to environmental stresses. Toxoplasma LEA-like genes (TgLEAs) exhibit the characteristic features of intrinsically disordered proteins, explaining some of their properties. Our in vitro biochemical experiments using recombinant TgLEA proteins show that they have cryoprotective effects on the oocyst-resident lactate dehydrogenase enzyme and that induced expression in E. coli of two of them leads to better survival after cold stress. Oocysts from a strain in which the four LEA genes were knocked out en bloc were significantly more susceptible to high salinity, freezing, and desiccation compared to wild-type oocysts. We discuss the evolutionary acquisition of LEA-like genes in Toxoplasma and other oocyst-producing apicomplexan parasites of the Sarcocystidae family and discuss how this has likely contributed to the ability of sporozoites within oocysts to survive outside the host for extended periods. Collectively, our data provide a first molecular detailed view on a mechanism that contributes to the remarkable resilience of oocysts against environmental stresses. IMPORTANCE Toxoplasma gondii oocysts are highly infectious and may survive in the environment for years. Their resistance against disinfectants and irradiation has been attributed to the oocyst and sporocyst walls by acting as physical and permeability barriers. However, the genetic basis for their resistance against stressors like changes in temperature, salinity, or humidity, is unknown. We show that a cluster of four genes encoding Toxoplasma Late Embryogenesis Abundant (TgLEA)-related proteins are important for this resistance to environmental stresses. TgLEAs have features of intrinsically disordered proteins, explaining some of their properties. Recombinant TgLEA proteins show cryoprotective effects on the parasite's lactate dehydrogenase, an abundant enzyme in oocysts, and expression in E. coli of two TgLEAs has a beneficial effect on growth after cold stress. Moreover, oocysts from a strain lacking all four TgLEA genes were more susceptible to high salinity, freezing, and desiccation compared to wild-type oocysts, highlighting the importance of the four TgLEAs for oocyst resilience.
Collapse
|
6
|
Bekier A, Brzostek A, Paneth A, Dziadek B, Dziadek J, Gatkowska J, Dzitko K. 4-Arylthiosemicarbazide Derivatives as Toxoplasmic Aromatic Amino Acid Hydroxylase Inhibitors and Anti-inflammatory Agents. Int J Mol Sci 2022; 23:ijms23063213. [PMID: 35328634 PMCID: PMC8955734 DOI: 10.3390/ijms23063213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/12/2022] Open
Abstract
Approximately one-third of the human population is infected with the intracellular cosmopolitan protozoan Toxoplasma gondii (Tg), and a specific treatment for this parasite is still needed. Additionally, the increasing resistance of Tg to drugs has become a challenge for numerous research centers. The high selectivity of a compound toward the protozoan, along with low cytotoxicity toward the host cells, form the basis for further research, which aims at determining the molecular targets of the active compounds. Thiosemicarbazide derivatives are biologically active organic compounds. Previous studies on the initial preselection of 58 new 4-arylthiosemicarbazide derivatives in terms of their anti-Tg activity and selectivity made it possible to select two promising derivatives for further research. One of the important amino acids involved in the proliferation of Tg and the formation of parasitophorous vacuoles is tyrosine, which is converted by two unique aromatic amino acid hydroxylases to levodopa. Enzymatic studies with two derivatives (R: para-nitro and meta-iodo) and recombinant aromatic amino acid hydroxylase (AAHs) obtained in the E. coli expression system were performed, and the results indicated that toxoplasmic AAHs are a molecular target for 4-arylthiosemicarbazide derivatives. Moreover, the drug affinity responsive target stability assay also confirmed that the selected compounds bind to AAHs. Additionally, the anti-inflammatory activity of these derivatives was tested using THP1-Blue™ NF-κB reporter cells due to the similarity of the thiosemicarbazide scaffold to thiosemicarbazone, both of which are known NF-κB pathway inhibitors.
Collapse
Affiliation(s)
- Adrian Bekier
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (A.B.); (B.D.); (J.G.)
| | - Anna Brzostek
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology, Polish Academy of Sciences, 93-232 Lodz, Poland; (A.B.); (J.D.)
| | - Agata Paneth
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Bożena Dziadek
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (A.B.); (B.D.); (J.G.)
| | - Jarosław Dziadek
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology, Polish Academy of Sciences, 93-232 Lodz, Poland; (A.B.); (J.D.)
| | - Justyna Gatkowska
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (A.B.); (B.D.); (J.G.)
| | - Katarzyna Dzitko
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (A.B.); (B.D.); (J.G.)
- Correspondence:
| |
Collapse
|
7
|
Nayeri T, Sarvi S, Daryani A. Toxoplasmosis: Targeting neurotransmitter systems in psychiatric disorders. Metab Brain Dis 2022; 37:123-146. [PMID: 34476718 DOI: 10.1007/s11011-021-00824-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/14/2021] [Indexed: 12/30/2022]
Abstract
The most common form of the disease caused by Toxoplasma gondii (T. gondii) is latent toxoplasmosis due to the formation of tissue cysts in various organs, such as the brain. Latent toxoplasmosis is probably a risk factor in the development of some neuropsychiatric disorders. Behavioral changes after infection are caused by the host immune response, manipulation by the parasite, central nervous system (CNS) inflammation, as well as changes in hormonal and neuromodulator relationships. The present review focused on the exact mechanisms of T. gondii effect on the alteration of behavior and neurotransmitter levels, their catabolites and metabolites, as well as the interaction between immune responses and this parasite in the etiopathogenesis of psychiatric disorders. The dysfunction of neurotransmitters in the neural transmission is associated with several neuropsychiatric disorders. However, further intensive studies are required to determine the effect of this parasite on altering the level of neurotransmitters and the role of neurotransmitters in the etiology of host behavioral changes.
Collapse
Affiliation(s)
- Tooran Nayeri
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahabeddin Sarvi
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Daryani
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
8
|
Global phosphoproteome analysis reveals significant differences between sporulated oocysts of virulent and avirulent strains of Toxoplasma gondii. Microb Pathog 2021; 161:105240. [PMID: 34655729 DOI: 10.1016/j.micpath.2021.105240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/15/2021] [Accepted: 10/06/2021] [Indexed: 11/23/2022]
Abstract
In this study, the differences in the phosphoproteomic landscape of sporulated oocysts between virulent and avirulent strains of Toxoplasma gondii were examined using a global phosphoproteomics approach. Phosphopeptides from sporulated oocysts of the virulent PYS strain (Chinese ToxoDB#9) and the avirulent PRU strain (type II) were enriched by titanium dioxide (TiO2) affinity chromatography and quantified using IBT approach. A total of 10,645 unique phosphopeptides, 8181 nonredundant phosphorylation sites and 2792 phosphoproteins were identified. We also detected 4129 differentially expressed phosphopeptides (DEPs) between sporulated oocysts of PYS strain and PRU strain (|log1.5 fold change| > 1 and p < 0.05), including 2485 upregulated and 1644 downregulated phosphopeptides. Motif analysis identified 24 motifs from the upregulated phosphorylated peptides including 22 serine motifs and two threonine motifs (TPE and TP), and 15 motifs from the downregulated phosphorylated peptides including 12 serine motifs and three threonine motifs (TP, RxxT and KxxT) in PYS strain when comparing PYS strain to PRU strain. Several kinases were consistent with motifs of overrepresented phosphopeptides, such as PKA, PKG, CKII, IKK, MAPK, EGFR, INSR, Jak, Syk, Src, Ab1. GO enrichment, KEGG pathway analysis and STRING analysis revealed DEPs significantly enriched in many biological processes and pathways. Kinase related network analysis showed that AGC kinase was the most connected kinase peptide. Our findings reveal significant difference in phosphopeptide profiles of sporulated oocysts between virulent and avirulent T. gondii strains, providing new resources for further elucidation of the mechanisms underpinning the virulence of T. gondii.
Collapse
|
9
|
Acute toxoplasmosis can increase serum dopamine level. J Parasit Dis 2021; 46:337-342. [DOI: 10.1007/s12639-021-01447-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/29/2021] [Indexed: 10/20/2022] Open
|
10
|
Bekier A, Węglińska L, Paneth A, Paneth P, Dzitko K. 4-Arylthiosemicarbazide derivatives as a new class of tyrosinase inhibitors and anti- Toxoplasma gondii agents. J Enzyme Inhib Med Chem 2021; 36:1145-1164. [PMID: 34074198 PMCID: PMC8174488 DOI: 10.1080/14756366.2021.1931164] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
We report herein anti-proliferation effects of 4-arylthiosemicarbazides, with a cyclopentane substitution at N1 position, on highly virulent RH strain of Toxoplasma gondii. Among them, the highest in vitro anti-Toxoplasma activity was found with the meta-iodo derivative. Further experiments demonstrated inhibitory effects of thiosemicarbazides on tyrosinase (Tyr) activity, and good correlation was found between percentage of Tyr inhibition and IC50Tg. To confirm the concept that thiosemicarbazides are able to disrupt tyrosine metabolism in Toxoplasma tachyzoites, the most potent Tyr inhibitors were tested for their efficacy of T. gondii growth inhibition. All of them significantly reduced the number of tachyzoites in the parasitophorous vacuoles (PVs) compared to untreated cells, as well as inhibited tachyzoites growth by impeding cell division. Collectively, these results indicate that compounds with the thiosemicarbazide scaffold are able to disrupt tyrosine metabolism in Toxoplasma tachyzoites by deregulation of their crucial enzyme tyrosine hydroxylase (TyrH).
Collapse
Affiliation(s)
- Adrian Bekier
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Lidia Węglińska
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Agata Paneth
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Piotr Paneth
- Institute of Applied Radiation Chemistry, Lodz University of Technology, Lodz, Poland.,Institute Center for Research on Innovative Biobased Materials (ICRI-BioM) - International Research Agenda, Lodz University of Technology, Lodz, Poland
| | - Katarzyna Dzitko
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| |
Collapse
|
11
|
Hatam-Nahavandi K, Calero-Bernal R, Rahimi MT, Pagheh AS, Zarean M, Dezhkam A, Ahmadpour E. Toxoplasma gondii infection in domestic and wild felids as public health concerns: a systematic review and meta-analysis. Sci Rep 2021; 11:9509. [PMID: 33947922 PMCID: PMC8097069 DOI: 10.1038/s41598-021-89031-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 04/13/2021] [Indexed: 02/02/2023] Open
Abstract
Felidae as definitive hosts for Toxoplasma gondii play a major role in transmission to all warm-blooded animals trough oocysts dissemination. Therefore the current comprehensive study was performed to determine the global status of T. gondii infection in domestic and wild felids aiming to provide comprehensive data of interest for further intervention approaching the One Health perspective. Different databases were searched by utilizing particular key words for publications related to T. gondii infecting domestic and wild feline host species, worldwide, from 1970 to 2020. The review of 337 reports showed that the seroprevalence of T. gondii in domestic cats and wild felids was estimated in 37.5% (95% CI 34.7-40.3) (I2 = 98.3%, P < 0.001) and 64% (95% CI 60-67.9) (I2 = 88%, P < 0.0001), respectively. The global pooled prevalence of oocysts in the fecal examined specimens from domestic cats was estimated in 2.6% (95% CI 1.9-3.3) (I2 = 96.1%, P < 0.0001), and that in fecal samples from wild felids was estimated in 2.4% (95% CI 1.1-4.2) (I2 = 86.4%, P < 0.0001). In addition, from 13,252 examined soil samples in 14 reviewed studies, the pooled occurrence of T. gondii oocysts was determined in 16.2% (95% CI 7.66-27.03%). The observed high rates of anti-T. gondii antibodies seroprevalence levels and oocyst excretion frequency in the felids, along with soil (environmental) contamination with oocysts may constitute a potential threat to animal and public health, and data will result of interest in further prophylaxis programs.
Collapse
Affiliation(s)
| | - Rafael Calero-Bernal
- grid.4795.f0000 0001 2157 7667SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| | - Mohammad Taghi Rahimi
- grid.444858.10000 0004 0384 8816Center for Health Related Social and Behavioral Sciences Research, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Abdol Sattar Pagheh
- grid.411701.20000 0004 0417 4622Infectious Diseases Research Canter, Birjand University of Medical Sciences, Birjand, Iran
| | - Mehdi Zarean
- grid.411583.a0000 0001 2198 6209Department of Parasitology and Mycology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asiyeh Dezhkam
- School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Ehsan Ahmadpour
- grid.412888.f0000 0001 2174 8913Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran ,grid.412888.f0000 0001 2174 8913Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran ,grid.412888.f0000 0001 2174 8913Department of Parasitology and Mycology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
12
|
Krishnan A, Soldati-Favre D. Amino Acid Metabolism in Apicomplexan Parasites. Metabolites 2021; 11:61. [PMID: 33498308 PMCID: PMC7909243 DOI: 10.3390/metabo11020061] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/05/2021] [Accepted: 01/14/2021] [Indexed: 12/22/2022] Open
Abstract
Obligate intracellular pathogens have coevolved with their host, leading to clever strategies to access nutrients, to combat the host's immune response, and to establish a safe niche for intracellular replication. The host, on the other hand, has also developed ways to restrict the replication of invaders by limiting access to nutrients required for pathogen survival. In this review, we describe the recent advancements in both computational methods and high-throughput -omics techniques that have been used to study and interrogate metabolic functions in the context of intracellular parasitism. Specifically, we cover the current knowledge on the presence of amino acid biosynthesis and uptake within the Apicomplexa phylum, focusing on human-infecting pathogens: Toxoplasma gondii and Plasmodium falciparum. Given the complex multi-host lifecycle of these pathogens, we hypothesize that amino acids are made, rather than acquired, depending on the host niche. We summarize the stage specificities of enzymes revealed through transcriptomics data, the relevance of amino acids for parasite pathogenesis in vivo, and the role of their transporters. Targeting one or more of these pathways may lead to a deeper understanding of the specific contributions of biosynthesis versus acquisition of amino acids and to design better intervention strategies against the apicomplexan parasites.
Collapse
Affiliation(s)
- Aarti Krishnan
- Department of Microbiology and Molecular Medicine, University of Geneva, CMU, Rue Michel-Servet 1, 1211 Geneva, Switzerland;
| | | |
Collapse
|
13
|
Behavioral Manipulation by Toxoplasma gondii: Does Brain Residence Matter? Trends Parasitol 2021; 37:381-390. [PMID: 33461902 DOI: 10.1016/j.pt.2020.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 12/04/2020] [Accepted: 12/27/2020] [Indexed: 11/22/2022]
Abstract
The protozoan parasite Toxoplasma gondii infects a wide range of intermediate hosts. The parasite produces brain cysts during the latent phase of its infection, in parallel to causing a loss of innate aversion in the rat host towards cat odors. Host behavioral change presumably reflects a parasitic manipulation to increase predation by definitive felid hosts, although evidence for increased predation is not yet available. In this opinion piece, we propose a neuroendocrine loop to explain the role of gonadal steroids in the parasitized hosts in mediating the behavioral manipulation. We argue that the presence of tissue cysts within the host brain is merely incidental to the behavioral change, without a necessary or sufficient role.
Collapse
|
14
|
Martorelli Di Genova B, Knoll LJ. Comparisons of the Sexual Cycles for the Coccidian Parasites Eimeria and Toxoplasma. Front Cell Infect Microbiol 2020; 10:604897. [PMID: 33381466 PMCID: PMC7768002 DOI: 10.3389/fcimb.2020.604897] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Toxoplasma gondii and Eimeria spp. are widely prevalent Coccidian parasites that undergo sexual reproduction during their life cycle. T. gondii can infect any warm-blooded animal in its asexual cycle; however, its sexual cycle is restricted to felines. Eimeria spp. are usually restricted to one host species, and their whole life cycle is completed within this same host. The literature reviewed in this article comprises the recent findings regarding the unique biology of the sexual development of T. gondii and Eimeria spp. The molecular basis of sex in these pathogens has been significantly unraveled by new findings in parasite differentiation along with transcriptional analysis of T. gondii and Eimeria spp. pre-sexual and sexual stages. Focusing on the metabolic networks, analysis of these transcriptome datasets shows enrichment for several different metabolic pathways. Transcripts for glycolysis enzymes are consistently more abundant in T. gondii cat infection stages than the asexual tachyzoite stage and Eimeria spp. merozoite and gamete stages compared to sporozoites. Recent breakthroughs in host-pathogen interaction and host restriction have significantly expanded the understating of the unique biology of these pathogens. This review aims to critically explore advances in the sexual cycle of Coccidia parasites with the ultimate goal of comparing and analyzing the sexual cycle of Eimeria spp. and T. gondii.
Collapse
Affiliation(s)
| | - Laura J. Knoll
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
15
|
Carrillo GL, Ballard VA, Glausen T, Boone Z, Teamer J, Hinkson CL, Wohlfert EA, Blader IJ, Fox MA. Toxoplasma infection induces microglia-neuron contact and the loss of perisomatic inhibitory synapses. Glia 2020; 68:1968-1986. [PMID: 32157745 PMCID: PMC7423646 DOI: 10.1002/glia.23816] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/17/2022]
Abstract
Infection and inflammation within the brain induces changes in neuronal connectivity and function. The intracellular protozoan parasite, Toxoplasma gondii, is one pathogen that infects the brain and can cause encephalitis and seizures. Persistent infection by this parasite is also associated with behavioral alterations and an increased risk for developing psychiatric illness, including schizophrenia. Current evidence from studies in humans and mouse models suggest that both seizures and schizophrenia result from a loss or dysfunction of inhibitory synapses. In line with this, we recently reported that persistent T. gondii infection alters the distribution of glutamic acid decarboxylase 67 (GAD67), an enzyme that catalyzes GABA synthesis in inhibitory synapses. These changes could reflect a redistribution of presynaptic machinery in inhibitory neurons or a loss of inhibitory nerve terminals. To directly assess the latter possibility, we employed serial block face scanning electron microscopy (SBFSEM) and quantified inhibitory perisomatic synapses in neocortex and hippocampus following parasitic infection. Not only did persistent infection lead to a significant loss of perisomatic synapses, it induced the ensheathment of neuronal somata by myeloid-derived cells. Immunohistochemical, genetic, and ultrastructural analyses revealed that these myeloid-derived cells included activated microglia. Finally, ultrastructural analysis identified myeloid-derived cells enveloping perisomatic nerve terminals, suggesting they may actively displace or phagocytose synaptic elements. Thus, these results suggest that activated microglia contribute to perisomatic inhibitory synapse loss following parasitic infection and offer a novel mechanism as to how persistent T. gondii infection may contribute to both seizures and psychiatric illness.
Collapse
Affiliation(s)
- Gabriela L. Carrillo
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, 2 Riverside Circle, Roanoke, VA 24016
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061
| | - Valerie A. Ballard
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, 2 Riverside Circle, Roanoke, VA 24016
- Roanoke Valley Governor’s School, Roanoke VA 24015
| | - Taylor Glausen
- Department of Microbiology and Immunology, University at Buffalo, Buffalo NY 14260
| | - Zack Boone
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, 2 Riverside Circle, Roanoke, VA 24016
- School of Neuroscience, Virginia Tech, Blacksburg, VA 24061
| | - Joseph Teamer
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, 2 Riverside Circle, Roanoke, VA 24016
- FBRI neuroSURF Program, Roanoke, VA 24016
| | - Cyrus L. Hinkson
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, 2 Riverside Circle, Roanoke, VA 24016
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016
| | | | - Ira J. Blader
- Department of Microbiology and Immunology, University at Buffalo, Buffalo NY 14260
| | - Michael A. Fox
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, 2 Riverside Circle, Roanoke, VA 24016
- School of Neuroscience, Virginia Tech, Blacksburg, VA 24061
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061
- Department of Pediatrics, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016
| |
Collapse
|
16
|
Zhao XY, Ewald SE. The molecular biology and immune control of chronic Toxoplasma gondii infection. J Clin Invest 2020; 130:3370-3380. [PMID: 32609097 PMCID: PMC7324197 DOI: 10.1172/jci136226] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Toxoplasma gondii is an incredibly successful parasite owing in part to its ability to persist within cells for the life of the host. Remarkably, at least 350 host species of T. gondii have been described to date, and it is estimated that 30% of the global human population is chronically infected. The importance of T. gondii in human health was made clear with the first reports of congenital toxoplasmosis in the 1940s. However, the AIDS crisis in the 1980s revealed the prevalence of chronic infection, as patients presented with reactivated chronic toxoplasmosis, underscoring the importance of an intact immune system for parasite control. In the last 40 years, there has been tremendous progress toward understanding the biology of T. gondii infection using rodent models, human cell experimental systems, and clinical data. However, there are still major holes in our understanding of T. gondii biology, including the genes controlling parasite development, the mechanisms of cell-intrinsic immunity to T. gondii in the brain and muscle, and the long-term effects of infection on host homeostasis. The need to better understand the biology of chronic infection is underscored by the recent rise in ocular disease associated with emerging haplotypes of T. gondii and our lack of effective treatments to sterilize chronic infection. This Review discusses the cell types and molecular mediators, both host and parasite, that facilitate persistent T. gondii infection. We highlight the consequences of chronic infection for tissue-specific pathology and identify open questions in this area of host-Toxoplasma interactions.
Collapse
|
17
|
Dubey JP, Cerqueira-Cézar CK, Murata FHA, Kwok OCH, Yang YR, Su C. All about toxoplasmosis in cats: the last decade. Vet Parasitol 2020; 283:109145. [PMID: 32645556 DOI: 10.1016/j.vetpar.2020.109145] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 12/14/2022]
Abstract
Toxoplasma gondii infections are common in humans and animals worldwide. Toxoplasmosis continues to be of public health concern. Cats (domestic and wild felids) are the most important host in the epidemiology of toxoplasmosis because they are the only species that can excrete the environmentally resistant oocysts in feces. Cats can excrete millions of oocysts and a single cat can spread infection to many hosts. The present paper summarizes information on prevalence, persistence of infection, clinical signs, and diagnosis of T. gondii infections in domestic and wild cats for the past decade. Special emphasis is paid to genetic diversity of T. gondii isolates from cats. Review of literature indicates that a unique genotype (ToxoDB genotype #9 or Chinese 1) is widely prevalent in cats in China and it has been epidemiologically linked to outbreaks of clinical toxoplasmosis in pigs and deaths in humans in China; this genotype has rarely been detected in other countries. This review will be of interest to biologists, parasitologists, veterinarians, and public health workers.
Collapse
Affiliation(s)
- J P Dubey
- United States Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD 20705-2350, USA.
| | - C K Cerqueira-Cézar
- United States Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD 20705-2350, USA
| | - F H A Murata
- United States Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD 20705-2350, USA
| | - O C H Kwok
- United States Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD 20705-2350, USA
| | - Y R Yang
- Laboratory of Veterinary Pathology, College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, PR China
| | - C Su
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996-0845, USA
| |
Collapse
|
18
|
Identification of Toxoplasma Gondii Tyrosine Hydroxylase (TH) Activity and Molecular Immunoprotection against Toxoplasmosis. Vaccines (Basel) 2020; 8:vaccines8020158. [PMID: 32244791 PMCID: PMC7349186 DOI: 10.3390/vaccines8020158] [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: 02/29/2020] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 02/07/2023] Open
Abstract
The neurotropic parasite Toxoplasma gondii (T. gondii) infection can change the behavior of rodents and cause neuropsychological symptoms in humans, which may be related to the change in neurotransmitter dopamine in the host brain caused by T. gondii infection. T. gondii tyrosine hydroxylase (TgTH) is an important factor in increasing the neurotransmitter dopamine in the host brain. In this study, the enzyme activity of TgTH catalytic substrate for dopamine production and the molecular characteristics of TgTH were identified. In order to amplify the open reading frame (ORF), the designing of the specific primers for polymerase chain reaction (PCR) was on the basis of the TgTH sequence (GenBank Accession No. EU481510.1), which was inserted into pET-32a (+) for the expression of recombined TgTH (rTgTH). The sequence analysis indicated that the gene of TgTH directed the encoding of a 62.4-kDa protein consisting of 565 amino acid residues, which was predicted to have a high antigen index. The enzyme activity test showed that rTgTH and the soluble proteins extracted separately from T. gondii RH strain and PRU strain could catalyze the substrate to produce dopamine in a dose-dependent manner, and the optimum catalytic temperature was 37 °C. The result of the Western Blotting assay revealed that the rTgTH and the native TgTH extracted from somatic of T. gondii RH tachyzoite were successfully detected by the sera of mice infected with T. gondii and the rat serum after rTgTH immune, respectively. Immunofluorescence analysis using antibody against rTgTH demonstrated that the protein was expressed and located on the surface of T. gondii RH tachyzoite. Freund’s adjuvant was used to emulsify the rTgTH, which was subsequently applied to BALB/c mouse immune thrice on week 0, week 2, and week 4, respectively. The result of the animal challenge experiments showed an integral increase in IgG, IgG2a, IgG1, and IFN-γ, IL-4, and IL17 were as well significantly increased, and that the rTgTH vaccinated animals apparently had a prolonged survival time (14.30 ± 2.41) after infection with the RH strain of T. gondii compared with that of the non-vaccinated control animals, which died within 11 days. Additionally, in the rTgTH vaccination group, the number of brain cysts (1275 ± 224) significantly decreased (p < 0.05) compared to the blank control group (2375 ± 883), and the size of the brain cysts in the animals immunized with rTgTH vaccination was remarkably smaller than that of the control mice. All the findings prove that TgTH played an important role in increasing the neurotransmitter dopamine in the host brain and could be used as a vaccine candidate antigen to mediate cell-mediated and humoral immunity.
Collapse
|
19
|
Freppel W, Ferguson DJ, Shapiro K, Dubey JP, Puech PH, Dumètre A. Structure, composition, and roles of the Toxoplasma gondii oocyst and sporocyst walls. Cell Surf 2019; 5:100016. [PMID: 32743133 PMCID: PMC7389338 DOI: 10.1016/j.tcsw.2018.100016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 12/03/2018] [Accepted: 12/14/2018] [Indexed: 01/01/2023] Open
Abstract
Toxoplasma gondii is a coccidian parasite with the cat as its definitive host but any warm-blooded animal, including humans, may act as intermediate hosts. It has a worldwide distribution where it may cause acute and chronic toxoplasmosis. Infection can result from ingestion either of tissue cysts in infected meat of intermediate hosts or oocysts found in cat faeces via contaminated water or food. In this review, we highlight how the oocyst and sporocyst walls sustain the persistence and transmission of infective T. gondii parasites from terrestrial and aquatic environments to the host. We further discuss why targeting the oocyst wall structure and molecules may reduce the burden of foodborne and waterborne T. gondii infections.
Collapse
|
20
|
Rosada B, Bekier A, Cytarska J, Płaziński W, Zavyalova O, Sikora A, Dzitko K, Łączkowski KZ. Benzo[b]thiophene-thiazoles as potent anti-Toxoplasma gondii agents: Design, synthesis, tyrosinase/tyrosine hydroxylase inhibitors, molecular docking study, and antioxidant activity. Eur J Med Chem 2019; 184:111765. [PMID: 31629163 DOI: 10.1016/j.ejmech.2019.111765] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/21/2019] [Accepted: 10/06/2019] [Indexed: 02/04/2023]
Abstract
Synthesis and investigation of anti-Toxoplasma gondii activity of novel thiazoles containing benzo [b]thiophene moiety are presented. Among the derivatives, compound 3k with adamantyl group shows exceptionally high potency against Me49 strain with IC50 (8.74 μM) value which is significantly lower than the activity of trimethoprim (IC50 39.23 μM). In addition, compounds 3a, 3b and 3k showed significant activity against RH strain (IC50 51.88-83.49 μM). The results of the cytotoxicity evaluation showed that Toxoplasma gondii growth was inhibited at non-cytotoxic concentrations for the mammalian L929 fibroblast (CC30 ∼ 880 μM). The most active compound 3k showed tyrosinase inhibition effect, with IC50 value of 328.5 μM. The binding energies calculated for compounds 3a-3e, 3k are strongly correlated with the experimentally determined values of tyrosinase inhibition activity. Moreover, the binding energies corresponding to the same ligands and calculated for both tyrosinase and tyrosine hydroxylase are also correlated with each other, suggesting that tyrosinase inhibitors may also have an inhibitory effect on tyrosine hydroxylase. Compounds 3j and 3k have also very strong antioxidant activity (IC50 15.9 and 15.5 μM), respectively, which is ten times higher than well-known antioxidant BHT.
Collapse
Affiliation(s)
- Beata Rosada
- Department of Chemical Technology and Pharmaceuticals, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Jurasza 2, 85-089, Bydgoszcz, Poland
| | - Adrian Bekier
- Department of Immunoparasitology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237, Lodz, Poland
| | - Joanna Cytarska
- Department of Chemical Technology and Pharmaceuticals, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Jurasza 2, 85-089, Bydgoszcz, Poland
| | - Wojciech Płaziński
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, 30-239, Cracow, Poland
| | - Olga Zavyalova
- Department of Chemical Technology and Pharmaceuticals, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Jurasza 2, 85-089, Bydgoszcz, Poland
| | - Adam Sikora
- Department of Medicinal Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Jurasza 2, 85-089, Bydgoszcz, Poland
| | - Katarzyna Dzitko
- Department of Immunoparasitology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237, Lodz, Poland.
| | - Krzysztof Z Łączkowski
- Department of Chemical Technology and Pharmaceuticals, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Jurasza 2, 85-089, Bydgoszcz, Poland.
| |
Collapse
|
21
|
Wallbank BA, Dominicus CS, Broncel M, Legrave N, Kelly G, MacRae JI, Staines HM, Treeck M. Characterisation of the Toxoplasma gondii tyrosine transporter and its phosphorylation by the calcium-dependent protein kinase 3. Mol Microbiol 2019; 111:1167-1181. [PMID: 30402958 PMCID: PMC6488386 DOI: 10.1111/mmi.14156] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2018] [Indexed: 12/21/2022]
Abstract
Toxoplasma gondii parasites rapidly exit their host cell when exposed to calcium ionophores. Calcium-dependent protein kinase 3 (TgCDPK3) was previously identified as a key mediator in this process, as TgCDPK3 knockout (∆cdpk3) parasites fail to egress in a timely manner. Phosphoproteomic analysis comparing WT with ∆cdpk3 parasites revealed changes in the TgCDPK3-dependent phosphoproteome that included proteins important for regulating motility, but also metabolic enzymes, indicating that TgCDPK3 controls processes beyond egress. Here we have investigated a predicted direct target of TgCDPK3, ApiAT5-3, a putative transporter of the major facilitator superfamily, and show that it is rapidly phosphorylated at serine 56 after induction of calcium signalling. Conditional knockout of apiAT5-3 results in transcriptional upregulation of most ribosomal subunits, but no alternative transporters, and subsequent parasite death. Mutating the S56 to a non-phosphorylatable alanine leads to a fitness cost, suggesting that phosphorylation of this residue is beneficial, albeit not essential, for tyrosine import. Using a combination of metabolomics and heterologous expression, we confirmed a primary role in tyrosine import for ApiAT5-3. However, no significant differences in tyrosine import could be detected in phosphorylation site mutants showing that if tyrosine transport is affected by S56 phosphorylation, its regulatory role is subtle.
Collapse
Affiliation(s)
- Bethan A. Wallbank
- Signalling in Apicomplexan Parasites LaboratoryThe Francis Crick InstituteLondonUK
| | - Caia S. Dominicus
- Signalling in Apicomplexan Parasites LaboratoryThe Francis Crick InstituteLondonUK
| | - Malgorzata Broncel
- Signalling in Apicomplexan Parasites LaboratoryThe Francis Crick InstituteLondonUK
| | - Nathalie Legrave
- Metabolomics Science Technology PlatformThe Francis Crick InstituteLondonUK
| | - Gavin Kelly
- Bioinformatics and Biostatistics STPFrancis Crick Institute1 Midland RoadLondon NW1 1ATUK
| | - James I. MacRae
- Metabolomics Science Technology PlatformThe Francis Crick InstituteLondonUK
| | - Henry M. Staines
- Institute of Infection and ImmunitySt George’s, University of LondonLondonUK
| | - Moritz Treeck
- Signalling in Apicomplexan Parasites LaboratoryThe Francis Crick InstituteLondonUK
| |
Collapse
|
22
|
Bahadori ES, Sadraei J, Dalimi A. New prevalence surveillance of Toxoplasma gondii among rodents and stray cats by ELISA avidity and nested PCR methods, Northeast of Iran. J Parasit Dis 2019; 43:360-367. [PMID: 31406400 DOI: 10.1007/s12639-019-01099-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 02/18/2019] [Indexed: 11/24/2022] Open
Abstract
Rodents and stray cats are the sources of many parasitic infections including T. gondii, for other animals and human. Toxoplasmosis has a wide range of laboratory factors in its intermediate and definite hosts. Regarding the importance of rodents and stray cats as the hosts that spread the Toxoplasma gondii, it is necessary to obtain comprehensive information about these animals in the life cycle of T. gondii. The objective was to investigate the new prevalence of toxoplasmosis among target animals in Iran, using GRA6 gene in combinacion with ELISA avidity. In this study, 286 rodents and 210 stray cats were collected and their heart tissues extracted to obtain DNA, blood samples and IgG Ab of T.gondii parasite. We detected the positive tissue samples in our study by the nested-PCR method. Then, we examined T. gondii IgG ELISA avidity for assessment of toxoplasmosis among rodents and stray cats. This study, was conducted in January to March 2017, based on the prevalence study. The findings revealed that 246/286 (86.01%) of rodents and 180/210 (85.71%) of stray cats were positive by IgG ELISA avidity methods. moreover, 68 rodents samples and 38 stray cats samples were positive concerning the GRA6 Toxoplasma gene; and these positive samples were at intermediate levels for IgG avidity. We concluded that the new prevalence of toxoplasmosis among rodents and stray cats was at high levels, using the serologic method in Northeast of Iran and the results of quantitative ELISA avidity were as the same as those of the nested-PCR for detecting recent toxoplasmosis in these hosts.
Collapse
Affiliation(s)
- Ehsan Shariat Bahadori
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javid Sadraei
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abdolhosein Dalimi
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
23
|
Parker KER, Fairweather SJ, Rajendran E, Blume M, McConville MJ, Bröer S, Kirk K, van Dooren GG. The tyrosine transporter of Toxoplasma gondii is a member of the newly defined apicomplexan amino acid transporter (ApiAT) family. PLoS Pathog 2019; 15:e1007577. [PMID: 30742695 PMCID: PMC6386423 DOI: 10.1371/journal.ppat.1007577] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 02/22/2019] [Accepted: 01/12/2019] [Indexed: 12/20/2022] Open
Abstract
Apicomplexan parasites are auxotrophic for a range of amino acids which must be salvaged from their host cells, either through direct uptake or degradation of host proteins. Here, we describe a family of plasma membrane-localized amino acid transporters, termed the Apicomplexan Amino acid Transporters (ApiATs), that are ubiquitous in apicomplexan parasites. Functional characterization of the ApiATs of Toxoplasma gondii indicate that several of these transporters are important for intracellular growth of the tachyzoite stage of the parasite, which is responsible for acute infections. We demonstrate that the ApiAT protein TgApiAT5-3 is an exchanger for aromatic and large neutral amino acids, with particular importance for L-tyrosine scavenging and amino acid homeostasis, and that TgApiAT5-3 is critical for parasite virulence. Our data indicate that T. gondii expresses additional proteins involved in the uptake of aromatic amino acids, and we present a model for the uptake and homeostasis of these amino acids. Our findings identify a family of amino acid transporters in apicomplexans, and highlight the importance of amino acid scavenging for the biology of this important phylum of intracellular parasites. The Apicomplexa comprise a large number of parasitic protozoa that have obligate intracellular lifestyles and cause significant human and animal diseases, including malaria, cryptosporidiosis, toxoplasmosis, coccidiosis in poultry, and various cattle fevers. Apicomplexans must scavenge essential nutrients from their hosts in order to proliferate and cause disease, including a range of amino acids. The direct uptake of these nutrients is presumed to be mediated by transporter proteins located in the plasma membrane of intracellular stages, although the identities of these proteins are poorly defined. Using a combination of bioinformatic, genetic, cell biological, and physiological approaches, we have characterized an apicomplexan-specific family of plasma membrane-localized transporter proteins that we have called the Apicomplexan Amino acid Transporters (ApiATs). We show that TgApiAT5-3, a member of the family in the apicomplexan Toxoplasma gondii, is an exchanger for aromatic and large neutral amino acids. In particular, it is critical for uptake of tyrosine, and for parasite virulence in a mouse infection model. We conclude that ApiATs are a family of plasma membrane transporters that play crucial roles in amino acid scavenging by apicomplexan parasites.
Collapse
Affiliation(s)
- Kathryn E. R. Parker
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | | | - Esther Rajendran
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Martin Blume
- Department of Biochemistry and Molecular Biology and the Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, VIC, Australia
- Robert Koch Institute, Berlin, Germany
| | - Malcolm J. McConville
- Department of Biochemistry and Molecular Biology and the Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, VIC, Australia
| | - Stefan Bröer
- Research School of Biology, Australian National University, Canberra, ACT, Australia
- * E-mail: (GGVD); (KK); (SB)
| | - Kiaran Kirk
- Research School of Biology, Australian National University, Canberra, ACT, Australia
- * E-mail: (GGVD); (KK); (SB)
| | - Giel G. van Dooren
- Research School of Biology, Australian National University, Canberra, ACT, Australia
- * E-mail: (GGVD); (KK); (SB)
| |
Collapse
|
24
|
Ramakrishnan C, Maier S, Walker RA, Rehrauer H, Joekel DE, Winiger RR, Basso WU, Grigg ME, Hehl AB, Deplazes P, Smith NC. An experimental genetically attenuated live vaccine to prevent transmission of Toxoplasma gondii by cats. Sci Rep 2019; 9:1474. [PMID: 30728393 PMCID: PMC6365665 DOI: 10.1038/s41598-018-37671-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/11/2018] [Indexed: 12/20/2022] Open
Abstract
Almost any warm-blooded creature can be an intermediate host for Toxoplasma gondii. However, sexual reproduction of T. gondii occurs only in felids, wherein fertilisation of haploid macrogametes by haploid microgametes, results in diploid zygotes, around which a protective wall develops, forming unsporulated oocysts. Unsporulated oocysts are shed in the faeces of cats and meiosis gives rise to haploid sporozoites within the oocysts. These, now infectious, sporulated oocysts contaminate the environment as a source of infection for people and their livestock. RNA-Seq analysis of cat enteric stages of T. gondii uncovered genes expressed uniquely in microgametes and macrogametes. A CRISPR/Cas9 strategy was used to create a T. gondii strain that exhibits defective fertilisation, decreased fecundity and generates oocysts that fail to produce sporozoites. Inoculation of cats with this engineered parasite strain totally prevented oocyst excretion following infection with wild-type T. gondii, demonstrating that this mutant is an attenuated, live, transmission-blocking vaccine.
Collapse
Affiliation(s)
- Chandra Ramakrishnan
- Institute of Parasitology, University of Zürich, Winterthurerstrasse 266a, 8057, Zürich, Switzerland
| | - Simone Maier
- Institute of Parasitology, University of Zürich, Winterthurerstrasse 266a, 8057, Zürich, Switzerland
| | - Robert A Walker
- Institute of Parasitology, University of Zürich, Winterthurerstrasse 266a, 8057, Zürich, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Deborah E Joekel
- Institute of Parasitology, University of Zürich, Winterthurerstrasse 266a, 8057, Zürich, Switzerland
| | - Rahel R Winiger
- Institute of Parasitology, University of Zürich, Winterthurerstrasse 266a, 8057, Zürich, Switzerland
| | - Walter U Basso
- Institute of Parasitology, University of Zürich, Winterthurerstrasse 266a, 8057, Zürich, Switzerland
| | - Michael E Grigg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Adrian B Hehl
- Institute of Parasitology, University of Zürich, Winterthurerstrasse 266a, 8057, Zürich, Switzerland.
| | - Peter Deplazes
- Institute of Parasitology, University of Zürich, Winterthurerstrasse 266a, 8057, Zürich, Switzerland.
| | - Nicholas C Smith
- Research School of Biology, Australian National University, Canberra, ACT, 0200, Australia. .,School of Science and Health, Western Sydney University, Parramatta South Campus, Sydney, NSW, 2116, Australia.
| |
Collapse
|
25
|
Alsaady I, Tedford E, Alsaad M, Bristow G, Kohli S, Murray M, Reeves M, Vijayabaskar MS, Clapcote SJ, Wastling J, McConkey GA. Downregulation of the Central Noradrenergic System by Toxoplasma gondii Infection. Infect Immun 2019; 87:e00789-18. [PMID: 30510101 PMCID: PMC6346129 DOI: 10.1128/iai.00789-18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 11/15/2018] [Indexed: 12/17/2022] Open
Abstract
Toxoplasma gondii is associated with physiological effects in the host. Dysregulation of catecholamines in the central nervous system has previously been observed in chronically infected animals. In the study described here, the noradrenergic system was found to be suppressed with decreased levels of norepinephrine (NE) in brains of infected animals and in infected human and rat neural cells in vitro The mechanism responsible for the NE suppression was found to be downregulation of dopamine β-hydroxylase (DBH) gene expression, encoding the enzyme that synthesizes norepinephrine from dopamine, with downregulation observed in vitro and in infected brain tissue, particularly in the dorsal locus coeruleus/pons region. The downregulation was sex specific, with males expressing reduced DBH mRNA levels whereas females were unchanged. Rather, DBH expression correlated with estrogen receptor in the female rat brains for this estrogen-regulated gene. DBH silencing was not a general response of neurons to infection, as human cytomegalovirus did not downregulate DBH expression. The noradrenergic-linked behaviors of sociability and arousal were altered in chronically infected animals, with a high correlation between DBH expression and infection intensity. A decrease in DBH expression in noradrenergic neurons can elevate dopamine levels, which provides a possible explanation for mixed observations of changes in this neurotransmitter with infection. Decreased NE is consistent with the loss of coordination and motor impairments associated with toxoplasmosis. Further, the altered norepinephrine synthesis observed here may, in part, explain behavioral effects of infection and associations with mental illness.
Collapse
Affiliation(s)
- Isra Alsaady
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Ellen Tedford
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Mohammad Alsaad
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Greg Bristow
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Shivali Kohli
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Matthew Murray
- Institute of Immunity & Transplantation, UCL Department of Virology, Division of Infection & Immunity, Royal Free Hospital, London, United Kingdom
| | - Matthew Reeves
- Institute of Immunity & Transplantation, UCL Department of Virology, Division of Infection & Immunity, Royal Free Hospital, London, United Kingdom
| | - M S Vijayabaskar
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Steven J Clapcote
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Jonathan Wastling
- Faculty of Natural Sciences, University of Keele, Newcastle-under-Lyme, United Kingdom
| | - Glenn A McConkey
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
26
|
Tyebji S, Seizova S, Hannan AJ, Tonkin CJ. Toxoplasmosis: A pathway to neuropsychiatric disorders. Neurosci Biobehav Rev 2018; 96:72-92. [PMID: 30476506 DOI: 10.1016/j.neubiorev.2018.11.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/23/2018] [Accepted: 11/22/2018] [Indexed: 12/24/2022]
Abstract
Toxoplasma gondii is an obligate intracellular parasite that resides, in a latent form, in the human central nervous system. Infection with Toxoplasma drastically alters the behaviour of rodents and is associated with the incidence of specific neuropsychiatric conditions in humans. But the question remains: how does this pervasive human pathogen alter behaviour of the mammalian host? This fundamental question is receiving increasing attention as it has far reaching public health implications for a parasite that is very common in human populations. Our current understanding centres on neuronal changes that are elicited directly by this intracellular parasite versus indirect changes that occur due to activation of the immune system within the CNS, or a combination of both. In this review, we explore the interactions between Toxoplasma and its host, the proposed mechanisms and consequences on neuronal function and mental health, and discuss Toxoplasma infection as a public health issue.
Collapse
Affiliation(s)
- Shiraz Tyebji
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, 3052, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, Victoria, Australia.
| | - Simona Seizova
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, 3052, Australia.
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, Victoria, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Parkville, 3052, Victoria, Australia.
| | - Christopher J Tonkin
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, 3052, Australia.
| |
Collapse
|
27
|
Xiao J, Prandovszky E, Kannan G, Pletnikov MV, Dickerson F, Severance EG, Yolken RH. Toxoplasma gondii: Biological Parameters of the Connection to Schizophrenia. Schizophr Bull 2018; 44:983-992. [PMID: 29889280 PMCID: PMC6101499 DOI: 10.1093/schbul/sby082] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
It is increasingly evident that the brain is not truly an immune privileged site and that cells of the central nervous system are sensitive to the inflammation generated when the brain is fighting off infection. Among the many microorganisms that have access to the brain, the apicomplexan protozoan Toxoplasma gondii has been one of the most studied. This parasite has been associated with many neuropsychiatric disorders including schizophrenia. This article provides a comprehensive review of the status of Toxoplasma research in schizophrenia. Areas of interest include (1) the limitations and improvements of immune-based assays to detect these infections in humans, (2) recent discoveries concerning the schizophrenia-Toxoplasma association, (3) findings of Toxoplasma neuropathology in animal models related to schizophrenia pathogenesis, (4) interactions of Toxoplasma with the host genome, (5) gastrointestinal effects of Toxoplasma infections, and (6) therapeutic intervention of Toxoplasma infections.
Collapse
Affiliation(s)
- Jianchun Xiao
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - Emese Prandovszky
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - Geetha Kannan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Mikhail V Pletnikov
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Faith Dickerson
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD
| | - Emily G Severance
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD,To whom correspondence should be addressed; Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21287 USA; tel: +1-410-614-0004, fax: +1-410-955-3723, e-mail:
| |
Collapse
|
28
|
McFarland R, Wang ZT, Jouroukhin Y, Li Y, Mychko O, Coppens I, Xiao J, Jones-Brando L, Yolken RH, Sibley LD, Pletnikov MV. AAH2 gene is not required for dopamine-dependent neurochemical and behavioral abnormalities produced by Toxoplasma infection in mouse. Behav Brain Res 2018; 347:193-200. [PMID: 29555339 DOI: 10.1016/j.bbr.2018.03.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/10/2018] [Accepted: 03/14/2018] [Indexed: 01/25/2023]
Abstract
Infection with the protozoan parasite, Toxoplasma gondii (T. gondii), has been associated with the increased risk for several psychiatric disorders. The exact mechanisms of a hypothesized contribution of T. gondii infection are poorly understood. The T. gondii genome contains two aromatic amino acid hydroxylase genes (AAH1 and AAH2) that encode proteins that can produce L-DOPA. One popular hypothesis posits that these encoded enzymes might influence dopamine (DA) production and hence DA synaptic transmission, leading to neurobehavioral abnormalities in the infected host. Prior studies have shown that deletion of these genes does not alter DA levels in the brain or exploratory activity in infected mice. However, possible effects of AAH gene deficiency on infection-induced brain and behavior alterations that are directly linked to DA synaptic transmission have not been evaluated. We found that chronic T. gondii infection of BALB/c mice leads to blunted response to amphetamine or cocaine and decreased expression of Dopamine Transporter (DAT) and Vesicular Monoamine Transporter 2 (VMAT2). Deletion of AAH2 had no effects on these changes in infected mice. Both wild type and Δaah2 strains produced comparable levels of neuroinflammation. Our findings demonstrate that AAH2 is not required for T. gondii infection-produced DA-dependent neurobehavioral abnormalities.
Collapse
Affiliation(s)
- Ross McFarland
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Zi Teng Wang
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yan Jouroukhin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Ye Li
- Stanley Neurovirology Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Olga Mychko
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Isabelle Coppens
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Jianchun Xiao
- Stanley Neurovirology Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Lorraine Jones-Brando
- Stanley Neurovirology Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Robert H Yolken
- Stanley Neurovirology Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Mikhail V Pletnikov
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
29
|
Insights into the molecular basis of host behaviour manipulation by Toxoplasma gondii infection. Emerg Top Life Sci 2017; 1:563-572. [PMID: 33525856 DOI: 10.1042/etls20170108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/22/2017] [Accepted: 11/27/2017] [Indexed: 12/22/2022]
Abstract
Typically illustrating the 'manipulation hypothesis', Toxoplasma gondii is widely known to trigger sustainable behavioural changes during chronic infection of intermediate hosts to enhance transmission to its feline definitive hosts, ensuring survival and dissemination. During the chronic stage of infection in rodents, a variety of neurological dysfunctions have been unravelled and correlated with the loss of cat fear, among other phenotypic impacts. However, the underlying neurological alteration(s) driving these behavioural modifications is only partially understood, which makes it difficult to draw more than a correlation between T. gondii infection and changes in brain homeostasis. Moreover, it is barely known which among the brain regions governing fear and stress responses are preferentially affected during T. gondii infection. Studies aiming at an in-depth dissection of underlying molecular mechanisms occurring at the host and parasite levels will be discussed in this review. Addressing this reminiscent topic in the light of recent technical progress and new discoveries regarding fear response, olfaction and neuromodulator mechanisms could contribute to a better understanding of this complex host-parasite interaction.
Collapse
|
30
|
Ablation of an Ovarian Tumor Family Deubiquitinase Exposes the Underlying Regulation Governing the Plasticity of Cell Cycle Progression in Toxoplasma gondii. mBio 2017; 8:mBio.01846-17. [PMID: 29162714 PMCID: PMC5698556 DOI: 10.1128/mbio.01846-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The Toxoplasma genome encodes the capacity for distinct architectures underlying cell cycle progression in a life cycle stage-dependent manner. Replication in intermediate hosts occurs by endodyogeny, whereas a hybrid of schizogony and endopolygeny occurs in the gut of the definitive feline host. Here, we characterize the consequence of the loss of a cell cycle-regulated ovarian tumor (OTU family) deubiquitinase, OTUD3A of Toxoplasma gondii (TgOTUD3A; TGGT1_258780), in T. gondii tachyzoites. Rather than the mutation being detrimental, mutant parasites exhibited a fitness advantage, outcompeting the wild type. This phenotype was due to roughly one-third of TgOTUD3A-knockout (TgOTUD3A-KO) tachyzoites exhibiting deviations from endodyogeny by employing replication strategies that produced 3, 4, or 5 viable progeny within a gravid mother instead of the usual 2. We established the mechanistic basis underlying these altered replication strategies to be a dysregulation of centrosome duplication, causing a transient loss of stoichiometry between the inner and outer cores that resulted in a failure to terminate S phase at the attainment of 2N ploidy and/or the decoupling of mitosis and cytokinesis. The resulting dysregulation manifested as deviations in the normal transitions from S phase to mitosis (S/M) (endopolygeny-like) or M phase to cytokinesis (M/C) (schizogony-like). Notably, these imbalances are corrected prior to cytokinesis, resulting in the generation of normal progeny. Our findings suggest that decisions regarding the utilization of specific cell cycle architectures are controlled by a ubiquitin-mediated mechanism that is dependent on the absolute threshold levels of an as-yet-unknown target(s). Analysis of the TgOTUD3A-KO mutant provides new insights into mechanisms underlying the plasticity of apicomplexan cell cycle architecture. Replication by Toxoplasma gondii can occur by 3 distinct cell cycle architectures. Endodyogeny is used by asexual stages, while a hybrid of schizogony and endopolygeny is used by merozoites in the definitive feline host. Here, we establish that the disruption of an ovarian-tumor (OTU) family deubiquitinase, TgOTUD3A, in tachyzoites results in dysregulation of the mechanism controlling the selection of replication strategy in a subset of parasites. The mechanistic basis for these altered cell cycles lies in the unique biology of the bipartite centrosome that is associated with the transient loss of stoichiometry between the inner and outer centrosome cores in the TgOTUD3A-KO mutant. This highlights the importance of ubiquitin-mediated regulation in the transition from the nuclear to the budding phases of the cell cycle and provides new mechanistic insights into the regulation of the organization of the apicomplexan cell cycle.
Collapse
|
31
|
Afonso C, Paixão VB, Klaus A, Lunghi M, Piro F, Emiliani C, Di Cristina M, Costa RM. Toxoplasma-induced changes in host risk behaviour are independent of parasite-derived AaaH2 tyrosine hydroxylase. Sci Rep 2017; 7:13822. [PMID: 29062106 PMCID: PMC5653819 DOI: 10.1038/s41598-017-13229-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/20/2017] [Indexed: 01/21/2023] Open
Abstract
Toxoplasma gondii infects a broad range of hosts and can establish chronic infections with the formation of brain cysts. Infected animals show altered risk behaviour which has been suggested to increase capture probability of hosts, and thus enhance parasite transmission. It has been proposed that the ability of Toxoplasma cysts to secrete tyrosine hydroxylase could mediate these behavioural alterations. We tested the involvement of secreted tyrosine hydroxylase, coded by the parasite AaaH2 gene, in the development of alterations in mouse behaviour, by generating an AaaH2 deletion mutant parasite strain and testing its influence on behaviour. We found that both mice infected with wild type or AaaH2 mutant strains showed changes in risk behaviour. We confirmed these findings using factor analysis of the behaviour, which revealed that behavioural changes happened along a single dimension, and were observed in both infected groups. Furthermore, we developed a new behavioural paradigm in which animals are unpredictably trapped, and observed that both groups of infected animals perceive trapping but fail to adjust their behaviour to avoid further trapping. These results demonstrate that parasite-secreted AaaH2 TH is neither necessary for the generation of risky behaviour nor for the increased trappability observed during chronic Toxoplasma infection.
Collapse
Affiliation(s)
- Cristina Afonso
- Champalimaud Center for the Unknown, Champalimaud Neuroscience Programme, Av. Brasília, Doca de Pedrouços, 1400-038, Lisboa, Portugal
| | - Vitor B Paixão
- Champalimaud Center for the Unknown, Champalimaud Neuroscience Programme, Av. Brasília, Doca de Pedrouços, 1400-038, Lisboa, Portugal
| | - Andreas Klaus
- Champalimaud Center for the Unknown, Champalimaud Neuroscience Programme, Av. Brasília, Doca de Pedrouços, 1400-038, Lisboa, Portugal
| | - Matteo Lunghi
- University of Perugia, Department of Chemistry, Biology and Biotechnology, Building B, Via del Giochetto, 06122, Perugia, Italy
| | - Federica Piro
- University of Perugia, Department of Chemistry, Biology and Biotechnology, Building B, Via del Giochetto, 06122, Perugia, Italy
| | - Carla Emiliani
- University of Perugia, Department of Chemistry, Biology and Biotechnology, Building B, Via del Giochetto, 06122, Perugia, Italy
| | - Manlio Di Cristina
- University of Perugia, Department of Chemistry, Biology and Biotechnology, Building B, Via del Giochetto, 06122, Perugia, Italy.
| | - Rui M Costa
- Champalimaud Center for the Unknown, Champalimaud Neuroscience Programme, Av. Brasília, Doca de Pedrouços, 1400-038, Lisboa, Portugal.
| |
Collapse
|