1
|
Johnson HJ, Kochanowsky JA, Chandrasekaran S, Hunter CA, Beiting DP, Koshy AA. Defining neuronal responses to the neurotropic parasite Toxoplasma gondii. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.645603. [PMID: 40236177 PMCID: PMC11996365 DOI: 10.1101/2025.03.31.645603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
A select group of pathogens infect neurons in the brain. Prior dogma held that neurons were "defenseless" against infecting microbes, but many studies suggest that neurons can mount anti-microbial defenses. However, a knowledge gap in understanding how neurons respond in vitro and in vivo to different classes of micro-organisms remains. To address this gap, we compared a transcriptional dataset derived from primary neuron cultures (PNCs) infected with the neurotropic intracellular parasite Toxoplasma gondii with a dataset derived from neurons injected with T. gondii protein in vivo . These curated responses were then compared to the transcriptional responses of PNCs infected with the single stranded RNA viruses West Nile Virus (WNV) or Zika Virus (ZKV). These analyses highlighted a conserved response to infection associated with chemokines ( Cxcl10, Ccl2 ) and cytokines (interferon signaling). However, T. gondii had diminished IFN-α signaling in vitro compared to the viral datasets and was uniquely associated with a decrease in neuron-specific genes ( Snap25 , Slc17a7 , Prkcg ). These data underscore that neurons participate in infection-induced neuroinflammation and illustrate that neurons possess both pathogen-specific and pathogen-conserved responses. Importance Though neurons are commonly the target of pathogens that infect the CNS, few datasets assess the neuronal response to infection. This paucity of data is likely because neurons are perceived to have diminished immune capabilities. However, to understand the role of neurons in neuroinflammation and their immune capabilities, their responses must be investigated. Here we analyzed publicly accessible, neuron-specific datasets to compare neuron responses to a eukaryotic pathogen versus two Orthoflaviviruses. A better understanding of neuron responses to different infections will allow us to develop methods for inhibiting pathways that lead to neuron dysfunction, enhancing those that limit pathogen survival, and mitigating infection-induced damage to the CNS.
Collapse
|
2
|
Ramírez-Flores CJ, Hryckowian ND, Gale AN, Babatunde KA, Lares M, Beebe DJ, Kerr SC, Knoll LJ. Modeling Toxoplasma gondii-gut early interactions using a human microphysiological system. PLoS Negl Trop Dis 2025; 19:e0012855. [PMID: 39903779 DOI: 10.1371/journal.pntd.0012855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
Oral transmission of parasites via environmentally resistant cyst stages in contaminated food or water is a common route of human infection, but there are no effective vaccines available for any enteric parasitic infection. Our knowledge of parasite cyst stage conversion and interaction with the intestinal tract is limited. Here, we investigate infection dynamics of Toxoplasma gondii cyst-stage in murine jejunum and human intestinal microphysiological systems. We focus on parasite ingress, replication, and conversion of the cyst stage to the rapidly replicating dissemination stage. In vivo bioluminescent imaging of mice fed cysts revealed spots of infection throughout the jejunum and ileum, which were selected for further analyses. Immunostaining showed parasite migration and replication predominantly in the stroma, with minimal replication in enterocytes. We recapitulated bradyzoite infection in human intestinal microphysiological systems and showed stage conversation and migration through collagen. This integrated approach elucidates complex host-parasite interactions, highlighting the value of microphysiological systems in advancing understanding and identifying potential therapeutics.
Collapse
Affiliation(s)
- Carlos J Ramírez-Flores
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Nicole D Hryckowian
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Andrew N Gale
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kehinde Adebayo Babatunde
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Marcos Lares
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Sheena C Kerr
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Laura J Knoll
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
3
|
Andreou D, Steen NE, Jørgensen KN, Ueland T, Wortinger LA, Mørch-Johnsen L, Drabløs I, Calkova T, Yolken RH, Andreassen OA, Agartz I. Increased Herpes simplex virus 1, Toxoplasma gondii and Cytomegalovirus antibody concentrations in severe mental illness. Transl Psychiatry 2024; 14:498. [PMID: 39695093 DOI: 10.1038/s41398-024-03198-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
Infections with Cytomegalovirus (CMV), Herpes simplex virus 1 (HSV1) and Toxoplasma gondii (TG) have been implicated in severe mental illness. All three pathogens have high seroprevalence in the human population, are neurotropic and establish a persistent infection. We hypothesized that exposed (seropositive) patients with severe mental illness would show higher immunoglobulin G (IgG) concentrations than exposed healthy controls (HC). We included 765 patients with severe mental illness (schizophrenia n = 515, bipolar disorder n = 250) and 541 HC. CMV, HSV1 and TG IgG seropositivity and concentrations were measured with immunoassays (seropositivity: CMV, n = 447 patients vs. 296 HC; HSV1, n = 355 vs. 238; and TG, n = 159 vs. 126). Among seropositive participants, patients had higher HSV1 (p < 0.001) and TG (p = 0.003) IgG concentrations than HC. Stratifying by diagnosis, both schizophrenia (p = 0.001) and bipolar disorder (p = 0.001) had higher HSV1 IgG concentrations, while schizophrenia only had higher TG (p = 0.009) and CMV (p = 0.045) IgG concentrations than HC. In SZ, higher HSV1 IgG concentrations were associated with higher psychotic (p = 0.030) and manic (p = 0.008) symptom scores, but only among CMV- or TG-infected patients which suggests synergistic effects. Among all participants, HSV1 IgG concentrations were inversely associated with interleukin-18 (p < 0.001) and positively associated with high-sensitivity C-reactive protein (p = 0.002) and B cell-activating factor (p = 0.004), possibly indicating T cell exhaustion, enhanced inflammation, and increased B-cell response, respectively. Patients with severe mental illness exhibit a heightened immune system response to HSV1, TG, and CMV infections suggesting immune system dysfunction and/or a more severe infection. For HSV1, higher IgG concentrations were linked to a greater clinical burden.
Collapse
Affiliation(s)
- Dimitrios Andreou
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway.
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden.
| | - Nils Eiel Steen
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Kjetil Nordbø Jørgensen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Thrombosis Research Center (TREC), Division of internal medicine, University hospital of North Norway, Tromsø, Norway
| | - Laura A Wortinger
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lynn Mørch-Johnsen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry & Department of Clinical Research, Østfold Hospital, Grålum, Norway
| | - Ina Drabløs
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tereza Calkova
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
- Region Vastmanland - Uppsala University, Centre for Clinical Research, Vastmanland Hospital Vasteras, Västerås, Sweden
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, US
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ingrid Agartz
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm Region, Stockholm, Sweden
| |
Collapse
|
4
|
Sleda MA, Pitafi ZF, Song W, Oldfield E, Moreno SNJ. Lipophilic bisphosphonates reduced cyst burden and ameliorated hyperactivity of mice chronically infected with Toxoplasma gondii. mBio 2024; 15:e0175624. [PMID: 39387586 PMCID: PMC11558998 DOI: 10.1128/mbio.01756-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/23/2024] [Indexed: 10/15/2024] Open
Abstract
The current treatments for toxoplasmosis are only active against fast-growing tachyzoites, present in acute infections, with little effect on slow-growing bradyzoites within tissue cysts, present in latent chronic infections. The mitochondrion of Toxoplasma gondii is essential for its survival, and one of the major anti-parasitic drugs, atovaquone, inhibits the mitochondrial electron transport chain at the coenzyme Q:cytochrome c oxidoreductase site. Coenzyme Q (also known as ubiquinone [UQ]) consists of a quinone head and a lipophilic, isoprenoid tail that anchors UQ to membranes. The synthesis of the isoprenoid unit is essential for cell growth and is inhibited by lipophilic bisphosphonates, which inhibit the parasite growth. In this work, we investigated the effect of lipophilic bisphosphonates on the chronic stages of T. gondii. We discovered that three lipophilic bisphosphonates (BPH-1218, BPH-1236, and BPH-1238), effective for the acute infection, were also effective in controlling the development of chronic stages. We showed effectiveness by testing them against in vitro cysts and in vivo derived tissue cysts and, most importantly, these compounds reduced the cyst burden in the brains of chronically infected mice. We monitored the activity of infected mice non-invasively and continuously with a novel device termed the CageDot. A decrease in activity accompanied the acute phase, but mice recovered to normal activity and showed signs of hyperactivity when the chronic infection was established. Moreover, treatment with atovaquone or BPH-1218 ameliorated the hyperactivity observed during the chronic infection.IMPORTANCETreatment for toxoplasmosis is challenged by a lack of effective drugs to eradicate the chronic stages. Most of the drugs currently used are poorly distributed to the central nervous system, and they trigger allergic reactions in a large number of patients. There is a compelling need for safe and effective treatments for toxoplasmosis. Bisphosphonates (BPs) are analogs of inorganic pyrophosphate and are used for the treatment of bone disorders. BPs target the isoprenoid pathway and are effective against several experimental parasitic infections. Some lipophilic BPs can specifically inhibit the mitochondrial activity of Toxoplasma gondii by interfering with the mechanism by which ubiquinone is inserted into the inner mitochondrial membrane. In this work, we present the effect of three lipophilic BPs against T. gondii chronic stages. We also present a new strategy for the monitoring of animal activity during disease and treatment that is non-invasive and continuous.
Collapse
Affiliation(s)
- Melissa A. Sleda
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Zaid F. Pitafi
- School of Electrical and Computer Engineering, University of Georgia, Athens, Georgia, USA
| | - WenZhan Song
- School of Electrical and Computer Engineering, University of Georgia, Athens, Georgia, USA
| | - Eric Oldfield
- Department of Chemistry, University of Illinois at Urbana Champaign, Urbana, Illinois, USA
| | - Silvia N. J. Moreno
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
5
|
Dey V, Holmes MJ, Bastos MS, Wek RC, Sullivan WJ. Cap-independent translation directs stress-induced differentiation of the protozoan parasite Toxoplasma gondii. J Biol Chem 2024; 300:107979. [PMID: 39542243 PMCID: PMC11697163 DOI: 10.1016/j.jbc.2024.107979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Translational control mechanisms modulate the microbial latency of eukaryotic pathogens, enabling them to evade immunity and drug treatments. The protozoan parasite Toxoplasma gondii persists in hosts by differentiating from proliferative tachyzoites to latent bradyzoites, which are housed inside tissue cysts. Transcriptional changes facilitating bradyzoite conversion are mediated by a Myb domain transcription factor called BFD1, whose mRNA is present in tachyzoites but not translated into protein until stress is applied to induce differentiation. We addressed the mechanisms by which translational control drives BFD1 synthesis in response to stress-induced parasite differentiation. Using biochemical and molecular approaches, we show that the 5'-leader of BFD1 mRNA is sufficient for preferential translation upon stress. The translational control of BFD1 mRNA is maintained when ribosome assembly near its 5'-cap is impaired by insertion of a 5'-proximal stem-loop and upon knockdown of the Toxoplasma cap-binding protein, eIF4E1. Moreover, we determined that a trans-acting RNA-binding protein called BFD2/ROCY1 is necessary for the cap-independent translation of BFD1 through its binding to the 5'-leader. Translation of BFD2 mRNA is also suggested to be preferentially induced under stress but by a cap-dependent mechanism. These results show that translational control and differentiation in Toxoplasma proceed through cap-independent mechanisms in addition to canonical cap-dependent translation. Our identification of cap-independent translation in protozoa underscores the antiquity of this mode of gene regulation in cellular evolution and its central role in stress-induced life-cycle events.
Collapse
Affiliation(s)
- Vishakha Dey
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael J Holmes
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Matheus S Bastos
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ronald C Wek
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - William J Sullivan
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
6
|
Vanagas L, Cristaldi C, La Bella G, Ganuza A, Angel SO, Alonso AM. A bioinformatic approach for the prediction and functional classification of Toxoplasma gondii long non-coding RNAs. Sci Rep 2024; 14:27687. [PMID: 39533086 PMCID: PMC11557611 DOI: 10.1038/s41598-024-79204-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as significant players in diverse cellular processes, including cell differentiation. Advancements in computational methodologies have facilitated the prediction of lncRNA functions, enabling insights even in non-model organisms like pathogenic parasites, in roles such as parasite development, antigenic variation, and epigenetics. In this work, we focus on the apicomplexan Toxoplasma gondii differentiation process, where the infective stage, tachyzoite, can develop into the cysted stage, bradyzoite, under stress conditions. Using a publicly available transcriptome dataset, we predicted putative lncRNA sequences associated with this differentiation process. Notably, a substantial proportion of these putative lncRNAs exhibited stage-specific expression, particularly at the bradyzoite stage. Furthermore, co-expression patterns between coding transcripts and putative TglncRNAs suggest their involvement in shared processes, such as bradyzoite development. Putative TglncRNA loci analysis revealed their potential influence on the expression of nearby coding genes, including subtelomeric genes unique to the T. gondii genome. Finally we propose a k-mer analysis approach to predict putative functional relationships between characterized lncRNAs from model organisms like Homo sapiens and the putative T. gondii lncRNAs. Our perspective led to predict putative T. gondii lncRNA that potentially could act mediating DNA damage repair pathways, opening a new study field to validate this kind of adaptive mechanisms of T. gondii in response to stress conditions.
Collapse
Affiliation(s)
- Laura Vanagas
- Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, Provincia de Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (UNSAM), San Martín, Argentina
| | - Constanza Cristaldi
- Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, Provincia de Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (UNSAM), San Martín, Argentina
| | - Gino La Bella
- Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, Provincia de Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (UNSAM), San Martín, Argentina
| | - Agustina Ganuza
- Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, Provincia de Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (UNSAM), San Martín, Argentina
| | - Sergio O Angel
- Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, Provincia de Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (UNSAM), San Martín, Argentina
| | - Andrés M Alonso
- Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, Provincia de Buenos Aires, Argentina.
- Escuela de Bio y Nanotecnologías (UNSAM), San Martín, Argentina.
| |
Collapse
|
7
|
Sundararajan A, Vora K, Saiyed S, Natesan S, Vaidya V. Association of Maternal Toxoplasma Gondii Molecular and Serological Positivity With Child's Gross-Motor Development and Behavior in Tribal Regions of Gujarat, India: A Prospective Study. Health Sci Rep 2024; 7:e70183. [PMID: 39540024 PMCID: PMC11558262 DOI: 10.1002/hsr2.70183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Affiliation(s)
| | - Kranti Vora
- Bagchi School of Public HealthAhmedabad UniversityAhmedabadGujaratIndia
| | | | | | - Vidhi Vaidya
- Institute of ScienceNirma UniversityAhmedabadGujaratIndia
| |
Collapse
|
8
|
Eom GD, Chu KB, Mao J, Yoon KW, Kang HJ, Moon EK, Kim SS, Quan FS. Heterologous immunization targeting the CST1 antigen confers better protection than ROP18 in mice. Nanomedicine (Lond) 2024; 19:2437-2446. [PMID: 39320318 PMCID: PMC11520538 DOI: 10.1080/17435889.2024.2403333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024] Open
Abstract
Aim: To evaluate the protective efficacy induced by heterologous immunization with recombinant baculoviruses or virus-like particles targeting the CST1 and ROP18 antigens of Toxoplasma gondii.Materials & methods: Recombinant baculovirus and virus-like particle vaccines expressing T. gondii CST1 or ROP18 antigens were developed to evaluate protective immunity in mice upon challenge infection with 450 Toxoplasma gondii (ME49).Results: Immunization with CST1 or ROP18 vaccines induced similar levels of T. gondii-specific IgG and IgA responses. Compared with ROP 18, CST1 vaccine showed better antibody-secreting cell response, germinal center B cell activation, and significantly reduced brain cyst burden and body weight loss.Conclusion: Our findings suggest that CST1 heterologous immunization elicited better protection than ROP18, providing important insight into improving the toxoplasmosis vaccine design strategy.
Collapse
Affiliation(s)
- Gi-Deok Eom
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ki Back Chu
- Department of Parasitology, Inje University College of Medicine, Busan, 47392, Republic of Korea
- Department of Infectious Disease & Malaria, Paik Institute of Clinical Research, Inje University, Busan, 47392, Republic of Korea
| | - Jie Mao
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Keon-Woong Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hae-Ji Kang
- Department of Microbiology, Dongguk University College of Medicine, Gyeongju, 38066, Republic of Korea
| | - Eun-Kyung Moon
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sung Soo Kim
- Medical Research Center for Bioreaction to Reactive Oxygen Species & Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Fu-Shi Quan
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Medical Research Center for Bioreaction to Reactive Oxygen Species & Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
9
|
Dey V, Holmes MJ, Bastos MS, Wek RC, Sullivan WJ. Cap-independent translation directs stress-induced differentiation of the protozoan parasite Toxoplasma gondii. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613578. [PMID: 39345546 PMCID: PMC11429880 DOI: 10.1101/2024.09.17.613578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Translational control mechanisms modulate microbial latency of eukaryotic pathogens, enabling them to evade immunity and drug treatments. The protozoan parasite Toxoplasma gondii persists in hosts by differentiating from proliferative tachyzoites to latent bradyzoites, which are housed inside tissue cysts. Transcriptional changes facilitating bradyzoite conversion are mediated by a Myb domain transcription factor called BFD1, whose mRNA is present in tachyzoites but not translated into protein until stress is applied to induce differentiation. We addressed the mechanisms by which translational control drives BFD1 synthesis in response to stress-induced parasite differentiation. Using biochemical and molecular approaches, we show that the 5'-leader of BFD1 mRNA is sufficient for preferential translation upon stress. The translational control of BFD1 mRNA is maintained when ribosome assembly near its 5'-cap is impaired by insertion of a 5'-proximal stem-loop and upon knockdown of the Toxoplasma cap-binding protein, eIF4E1. Moreover, we show that a trans -acting RNA-binding protein called BFD2/ROCY1 is necessary for cap-independent translation of BFD1 through its binding to the 5'-leader. Translation of BFD2 mRNA is also suggested to be preferentially induced under stress, but by a cap-dependent mechanism. These results show that translational control and differentiation in Toxoplasma proceed through cap-independent mechanisms in addition to canonical cap-dependent translation. Our identification of cap-independent translation in protozoa underscores the antiquity of this mode of gene regulation in cellular evolution and its central role in stress-induced life-cycle events.
Collapse
|
10
|
García-López LL, Vargas-Montes M, Osorio-Méndez JF, Cardona N, Hernández De Los Ríos A, Toro-Acevedo CA, Arenas-García JC, Mantilla-Muriel LE, Torres E, Valencia-Hernández JD, Acosta-Dávila A, de-la-Torre A, Celis-Giraldo D, Mejía Oquendo M, Sepúlveda-Arias JC, Gómez-Marín JE. CD8+ T-cell Exhaustion Phenotype in Human Asymptomatic and Ocular Toxoplasmosis. Ocul Immunol Inflamm 2024; 32:1218-1227. [PMID: 37315178 DOI: 10.1080/09273948.2023.2217906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/16/2023]
Abstract
This work analyzed exhaustion markers in CD8+ T-cell subpopulations in 21 samples of peripheral blood mononuclear cells (PBMCs) from individuals with ocular toxoplasmosis (n = 9), chronic asymptomatic toxoplasmosis (n = 7), and non-infected people (n = 5) by using RT-qPCR and flow cytometry techniques. The study found that gene expression of PD-1 and CD244, but not LAG-3, was higher in individuals with ocular toxoplasmosis versus individuals with asymptomatic infection or uninfected. Expression of PD1 in CD8+ central memory (CM) cells was higher in nine individuals with toxoplasmosis versus five uninfected individuals (p = .003). After ex vivo stimulation, an inverse correlation was found between the exhaustion markers and quantitative clinical characteristics (lesion size, recurrence index, and number of lesions). A total exhaustion phenotype was found in 55.5% (5/9) of individuals with ocular toxoplasmosis. Our results suggest that the CD8+ exhaustion phenotype is involved in the pathogenesis of ocular toxoplasmosis.
Collapse
Affiliation(s)
| | - Mónica Vargas-Montes
- GEPAMOL, Biomedical Research Center, Universidad del Quindío, Armenia, Quindío, Colombia
| | | | - Néstor Cardona
- GEPAMOL, Biomedical Research Center, Universidad del Quindío, Armenia, Quindío, Colombia
- Faculty of Dentistry, Universidad Antonio Nariño, Armenia, Quindío, Colombia
| | | | - Carlos Andrés Toro-Acevedo
- Grupo Infección e Inmunidad, Faculty of Health Sciences, Universidad Tecnológica de Pereira, Pereira, Colombia
| | | | - Luz Eliana Mantilla-Muriel
- Grupo Infección e Inmunidad, Faculty of Health Sciences, Universidad Tecnológica de Pereira, Pereira, Colombia
| | - Elizabeth Torres
- GEPAMOL, Biomedical Research Center, Universidad del Quindío, Armenia, Quindío, Colombia
| | | | | | - Alejandra de-la-Torre
- GEPAMOL, Biomedical Research Center, Universidad del Quindío, Armenia, Quindío, Colombia
- NeURos Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Daniel Celis-Giraldo
- GEPAMOL, Biomedical Research Center, Universidad del Quindío, Armenia, Quindío, Colombia
| | - Manuela Mejía Oquendo
- GEPAMOL, Biomedical Research Center, Universidad del Quindío, Armenia, Quindío, Colombia
| | - Juan Carlos Sepúlveda-Arias
- Grupo Infección e Inmunidad, Faculty of Health Sciences, Universidad Tecnológica de Pereira, Pereira, Colombia
| | | |
Collapse
|
11
|
Kou YJ, Gao J, Li R, Ma ZY, Elsheikha HM, Wu XJ, Zheng XN, Wang M, Zhu XQ. Functional Characterization of Six Eukaryotic Translation Initiation Factors of Toxoplasma gondii Using the CRISPR-Cas9 System. Int J Mol Sci 2024; 25:7834. [PMID: 39063076 PMCID: PMC11276994 DOI: 10.3390/ijms25147834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/07/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Eukaryotic translation initiation factors (eIFs) are crucial for initiating protein translation and ensuring the correct assembly of mRNA-ribosomal subunit complexes. In this study, we investigated the effects of deleting six eIFs in the apicomplexan parasite Toxoplasma gondii using the CRISPR-Cas9 system. We determined the subcellular localization of these eIFs using C-terminal endogenous tagging and immunofluorescence analysis. Four eIFs (RH::315150-6HA, RH::286090-6HA, RH::249370-6HA, and RH::211410-6HA) were localized in the cytoplasm, while RH::224235-6HA was localized in the apicoplast. Additionally, RH::272640-6HA was found in both the basal complex and the cytoplasm of T. gondii. Functional characterization of the six RHΔeIFs strains was conducted using plaque assay, cell invasion assay, intracellular growth assay and egress assay in vitro, and virulence assay in mice. Disruption of five eIF genes (RHΔ315150, RHΔ272640, RHΔ249370, RHΔ211410, and RHΔ224235) did not affect the ability of the T. gondii RH strain to invade, replicate, form plaques and egress in vitro, or virulence in Kunming mice (p > 0.05). However, the RHΔ286090 strain showed slightly reduced invasion efficiency and virulence (p < 0.01) compared to the other five RHΔeIFs strains and the wild-type strain. The disruption of the TGGT1_286090 gene significantly impaired the ability of tachyzoites to differentiate into bradyzoites in both type I RH and type II Pru strains. These findings reveal that the eukaryotic translation initiation factor TGGT1_286090 is crucial for T. gondii bradyzoite differentiation and may serve as a potential target for drug development and an attenuated vaccine against T. gondii.
Collapse
Affiliation(s)
- Yong-Jie Kou
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (Y.-J.K.); (J.G.); (Z.-Y.M.); (X.-J.W.); (X.-N.Z.)
- 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, China;
| | - Jin Gao
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (Y.-J.K.); (J.G.); (Z.-Y.M.); (X.-J.W.); (X.-N.Z.)
- 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, China;
| | - Rui Li
- 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, China;
| | - Zhi-Ya Ma
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (Y.-J.K.); (J.G.); (Z.-Y.M.); (X.-J.W.); (X.-N.Z.)
| | - Hany M. Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, Sutton Bonington Campus, University of Nottingham, Loughborough LE12 5RD, UK;
| | - Xiao-Jing Wu
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (Y.-J.K.); (J.G.); (Z.-Y.M.); (X.-J.W.); (X.-N.Z.)
- 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, China;
| | - Xiao-Nan Zheng
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (Y.-J.K.); (J.G.); (Z.-Y.M.); (X.-J.W.); (X.-N.Z.)
| | - Meng Wang
- 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, China;
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu 610213, China
| | - Xing-Quan Zhu
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (Y.-J.K.); (J.G.); (Z.-Y.M.); (X.-J.W.); (X.-N.Z.)
| |
Collapse
|
12
|
Diniz DG, de Oliveira JHP, Guerreiro LCF, de Menezes GC, de Assis ACL, Duarte TQ, dos Santos IBD, Maciel FD, Soares GLDS, Araújo SC, Franco FTDC, do Carmo EL, Morais RDAB, de Lima CM, Brites D, Anthony DC, Diniz JAP, Diniz CWP. Contrasting Disease Progression, Microglia Reactivity, Tolerance, and Resistance to Toxoplasma gondii Infection in Two Mouse Strains. Biomedicines 2024; 12:1420. [PMID: 39061995 PMCID: PMC11274029 DOI: 10.3390/biomedicines12071420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/02/2024] [Accepted: 06/08/2024] [Indexed: 07/28/2024] Open
Abstract
Our study investigated the innate immune response to Toxoplasma gondii infection by assessing microglial phenotypic changes and sickness behavior as inflammatory response markers post-ocular tachyzoite instillation. Disease progression in Swiss albino mice was compared with the previously documented outcomes in BALB/c mice using an identical ocular route and parasite burden (2 × 105 tachyzoites), with saline as the control. Contrary to expectations, the Swiss albino mice displayed rapid, lethal disease progression, marked by pronounced sickness behaviors and mortality within 11-12 days post-infection, while the survivors exhibited no apparent signs of infection. Comparative analysis revealed the T. gondii-infected BALB/c mice exhibited reduced avoidance of feline odors, while the infected Swiss albino mice showed enhanced avoidance responses. There was an important increase in microglial cells in the dentate gyrus molecular layer of the infected Swiss albino mice compared to the BALB/c mice and their respective controls. Hierarchical cluster and discriminant analyses identified three microglial morphological clusters, differentially affected by T. gondii infection across strains. The BALB/c mice exhibited increased microglial branching and complexity, while the Swiss albino mice showed reduced shrunken microglial arbors, diminishing their morphological complexity. These findings highlight strain-specific differences in disease progression and inflammatory regulation, indicating lineage-specific mechanisms in inflammatory responses, tolerance, and resistance. Understanding these elements is critical in devising control measures for toxoplasmosis.
Collapse
Affiliation(s)
- Daniel G. Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | - Jhonnathan H. P. de Oliveira
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Luma C. F. Guerreiro
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal do Pará, Campus Bragança, Bragança 68600-000, Pará, Brazil
| | - Gabriel C. de Menezes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Alexa C. L. de Assis
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Tainá Q. Duarte
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Izabelly B. D. dos Santos
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Flávia D. Maciel
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Gabrielly L. da S. Soares
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Sanderson C. Araújo
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Felipe T. de C. Franco
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Ediclei L. do Carmo
- Seção de Parasitologia, Instituto Evandro Chagas, Belém 67030-000, Pará, Brazil; (E.L.d.C.); (R.d.A.B.M.)
| | - Rafaela dos A. B. Morais
- Seção de Parasitologia, Instituto Evandro Chagas, Belém 67030-000, Pará, Brazil; (E.L.d.C.); (R.d.A.B.M.)
| | - Camila M. de Lima
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal;
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Daniel C. Anthony
- Laboratory of Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford OX1 2JD, UK;
| | - José A. P. Diniz
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Cristovam W. P. Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| |
Collapse
|
13
|
Holmes MJ, Bastos MS, Dey V, Severo V, Wek RC, Sullivan WJ. mRNA cap-binding protein eIF4E1 is a novel regulator of Toxoplasma gondii latency. mBio 2024; 15:e0295423. [PMID: 38747593 PMCID: PMC11237481 DOI: 10.1128/mbio.02954-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/15/2024] [Indexed: 05/28/2024] Open
Abstract
The protozoan parasite Toxoplasma gondii causes serious opportunistic disease due to its ability to persist in patients as latent tissue cysts. The molecular mechanisms coordinating conversion between proliferative parasites (tachyzoites) and latent cysts (bradyzoites) are not fully understood. We previously showed that phosphorylation of eIF2α accompanies bradyzoite formation, suggesting that this clinically relevant process involves regulation of mRNA translation. In this study, we investigated the composition and role of eIF4F multi-subunit complexes in translational control. Using CLIPseq, we find that the cap-binding subunit, eIF4E1, localizes to the 5'-end of all tachyzoite mRNAs, many of which show evidence of stemming from heterogeneous transcriptional start sites. We further show that eIF4E1 operates as the predominant cap-binding protein in two distinct eIF4F complexes. Using genetic and pharmacological approaches, we found that eIF4E1 deficiency triggers efficient spontaneous formation of bradyzoites without stress induction. Consistent with this result, we also show that stress-induced bradyzoites exhibit reduced eIF4E1 expression. Overall, our findings establish a novel role for eIF4F in translational control required for parasite latency and microbial persistence. IMPORTANCE Toxoplasma gondii is an opportunistic pathogen important to global human and animal health. There are currently no chemotherapies targeting the encysted form of the parasite. Consequently, a better understanding of the mechanisms controlling encystation is required. Here we show that the mRNA cap-binding protein, eIF4E1, regulates the encystation process. Encysted parasites reduce eIF4E1 levels, and depletion of eIF4E1 decreases the translation of ribosome-associated machinery and drives Toxoplasma encystation. Together, these data reveal a new layer of mRNA translational control that regulates parasite encystation and latency.
Collapse
Affiliation(s)
- Michael J Holmes
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Matheus S Bastos
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Vishakha Dey
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Vanessa Severo
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ronald C Wek
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - William J Sullivan
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
14
|
Nayeri T, Sarvi S, Daryani A. Effective factors in the pathogenesis of Toxoplasmagondii. Heliyon 2024; 10:e31558. [PMID: 38818168 PMCID: PMC11137575 DOI: 10.1016/j.heliyon.2024.e31558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/11/2024] [Accepted: 05/17/2024] [Indexed: 06/01/2024] Open
Abstract
Toxoplasma gondii (T. gondii) is a cosmopolitan protozoan parasite in humans and animals. It infects about 30 % of the human population worldwide and causes potentially fatal diseases in immunocompromised hosts and neonates. For this study, five English-language databases (ScienceDirect, ProQuest, Web of Science, PubMed, and Scopus) and the internet search engine Google Scholar were searched. This review was accomplished to draw a global perspective of what is known about the pathogenesis of T. gondii and various factors affecting it. Virulence and immune responses can influence the mechanisms of parasite pathogenesis and these factors are in turn influenced by other factors. In addition to the host's genetic background, the type of Toxoplasma strain, the routes of transmission of infection, the number of passages, and different phases of parasite life affect virulence. The identification of virulence factors of the parasite could provide promising insights into the pathogenesis of this parasite. The results of this study can be an incentive to conduct more intensive research to design and develop new anti-Toxoplasma agents (drugs and vaccines) to treat or prevent this infection. In addition, further studies are needed to better understand the key agents in the pathogenesis of T. gondii.
Collapse
Affiliation(s)
- Tooran Nayeri
- Infectious and Tropical Diseases Research Center, Dezful University of Medical Sciences, Dezful, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahabeddin Sarvi
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Daryani
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
15
|
Bando H, Murata Y, Han Y, Sugi T, Fukuda Y, Bzik DJ, Fox BA, Kato K. Toxoplasma gondii chitinase-like protein TgCLP1 regulates the parasite cyst burden. Front Cell Infect Microbiol 2024; 14:1359888. [PMID: 38828265 PMCID: PMC11140023 DOI: 10.3389/fcimb.2024.1359888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/23/2024] [Indexed: 06/05/2024] Open
Abstract
Toxoplasma, an important intracellular parasite of humans and animals, causes life-threatening toxoplasmosis in immunocompromised individuals. Although Toxoplasma secretory proteins during acute infection (tachyzoite, which divides rapidly and causes inflammation) have been extensively characterized, those involved in chronic infection (bradyzoite, which divides slowly and is surrounded by a cyst wall) remain uncertain. Regulation of the cyst wall is essential to the parasite life cycle, and polysaccharides, such as chitin, in the cyst wall are necessary to sustain latent infection. Toxoplasma secretory proteins during the bradyzoite stage may have important roles in regulating the cyst wall via polysaccharides. Here, we focused on characterizing the hypothetical T. gondii chitinase, chitinase-like protein 1 (TgCLP1). We found that the chitinase-like domain containing TgCLP1 is partially present in the bradyzoite microneme and confirmed, albeit partially, its previous identification in the tachyzoite microneme. Furthermore, although parasites lacking TgCLP1 could convert from tachyzoites to bradyzoites and make an intact cyst wall, they failed to convert from bradyzoites to tachyzoites, indicating that TgCLP1 is necessary for bradyzoite reactivation. Taken together, our findings deepen our understanding of the molecular basis of recrudescence and could contribute to the development of novel strategies for the control of toxoplasmosis.
Collapse
Affiliation(s)
- Hironori Bando
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Osaki, Miyagi, Japan
- Department of Parasitology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yuho Murata
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Yongmei Han
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Tatsuki Sugi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- Division of Collaboration and Education, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yasuhiro Fukuda
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Osaki, Miyagi, Japan
| | - David J. Bzik
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Barbara A. Fox
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Kentaro Kato
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Osaki, Miyagi, Japan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| |
Collapse
|
16
|
Quan JJ, Nikolov LA, Sha J, Wohlschlegel JA, Coppens I, Bradley PJ. Systematic characterization of all Toxoplasma gondii TBC domain-containing proteins identifies an essential regulator of Rab2 in the secretory pathway. PLoS Biol 2024; 22:e3002634. [PMID: 38713739 PMCID: PMC11101121 DOI: 10.1371/journal.pbio.3002634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 05/17/2024] [Accepted: 04/22/2024] [Indexed: 05/09/2024] Open
Abstract
Toxoplasma gondii resides in its intracellular niche by employing a series of specialized secretory organelles that play roles in invasion, host cell manipulation, and parasite replication. Rab GTPases are major regulators of the parasite's secretory traffic that function as nucleotide-dependent molecular switches to control vesicle trafficking. While many of the Rab proteins have been characterized in T. gondii, precisely how these Rabs are regulated remains poorly understood. To better understand the parasite's secretory traffic, we investigated the entire family of Tre2-Bub2-Cdc16 (TBC) domain-containing proteins, which are known to be involved in vesicle fusion and secretory protein trafficking. We first determined the localization of all 18 TBC domain-containing proteins to discrete regions of the secretory pathway or other vesicles in the parasite. Second, we use an auxin-inducible degron approach to demonstrate that the protozoan-specific TgTBC9 protein, which localizes to the endoplasmic reticulum (ER), is essential for parasite survival. Knockdown of TgTBC9 results in parasite growth arrest and affects the organization of the ER and mitochondrial morphology. TgTBC9 knockdown also results in the formation of large lipid droplets (LDs) and multi-membranous structures surrounded by ER membranes, further indicating a disruption of ER functions. We show that the conserved dual-finger active site in the TBC domain of the protein is critical for its GTPase-activating protein (GAP) function and that the Plasmodium falciparum orthologue of TgTBC9 can rescue the lethal knockdown. We additionally show by immunoprecipitation and yeast 2 hybrid analyses that TgTBC9 preferentially binds Rab2, indicating that the TBC9-Rab2 pair controls ER morphology and vesicular trafficking in the parasite. Together, these studies identify the first essential TBC protein described in any protozoan and provide new insight into intracellular vesicle trafficking in T. gondii.
Collapse
Affiliation(s)
- Justin J. Quan
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Lachezar A. Nikolov
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
| | - Jihui Sha
- Department of Biological Chemistry and Institute of Genomics and Proteomics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - James A. Wohlschlegel
- Department of Biological Chemistry and Institute of Genomics and Proteomics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Peter J. Bradley
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
17
|
Voyiatzaki C, Zare Chormizi AD, Tsoumani ME, Efstathiou A, Konstantinidis K, Chaniotis D, Chrysos G, Argyraki A, Papastamopoulos V, Kotsianopoulou M. Seroprevalence of Toxoplasma gondii among HIV Positive Patients under Surveillance in Greek Infectious Disease Units: A Screening Study with Comparative Evaluation of Serological Methods. Pathogens 2024; 13:375. [PMID: 38787227 PMCID: PMC11123859 DOI: 10.3390/pathogens13050375] [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: 03/29/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
This study aims to screen for IgG antibodies against Toxoplasma gondii (T. gondii) in the sera of 155 newly diagnosed Human Immunodeficiency Virus (HIV) positive patients under surveillance in Greek Infectious Disease Units. Additionally, risk factors based on patient demographics were examined, and a comparative evaluation of commercially available serological methods was conducted. Three methods were employed to detect IgG antibodies against T. gondii: Enzyme-Linked Immunosorbent Assay (ELISA), Indirect Immunofluorescence Antibody Test (IFAT), and Western Blot (WB), which was used as a reference here. Forty-nine sera samples were true-positive for IgG antibodies against T. gondii, resulting in a 31.61% positivity rate, and the immunoassay test statistical reliability analysis resulted in higher IFAT accuracy (90.97%) compared to ELISA (76.26%). Furthermore, statistical analysis of demographic and immunological data included in the study placed female and foreign/non-Greek individuals at 2.24 (p = 0.0009) and 2.34 (p = 0.0006) times higher risk of positive T. gondii IgG testing compared to their male and Greek counterparts, respectively. Our findings on positivity rates and comparative serology underscore the importance of early and suitable screening measures for newly diagnosed HIV+ patients to mitigate the life-threatening outcomes that may arise from a potential subsequent T. gondii activation.
Collapse
Affiliation(s)
- Chrysa Voyiatzaki
- Department of Biomedical Sciences, Division of Medical Laboratories Science, University of West Attica, 12243 Athens, Greece
| | - Apollon Dareios Zare Chormizi
- Department of Biomedical Sciences, Division of Medical Laboratories Science, University of West Attica, 12243 Athens, Greece
| | - Maria E. Tsoumani
- Department of Biomedical Sciences, Division of Medical Laboratories Science, University of West Attica, 12243 Athens, Greece
| | - Antonia Efstathiou
- Department of Biomedical Sciences, Division of Medical Laboratories Science, University of West Attica, 12243 Athens, Greece
- Immunology of Infection Group, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Konstantinos Konstantinidis
- Department of Medicine, Laboratory of Biology, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece
| | - Dimitrios Chaniotis
- Department of Biomedical Sciences, Division of Medical Laboratories Science, University of West Attica, 12243 Athens, Greece
| | - Georgios Chrysos
- Second Department of Internal Medicine, Tzaneio General Hospital of Piraeus, 18536 Athens, Greece
| | - Aikaterini Argyraki
- Department of Internal Medicine, Sotiria Thoracic Diseases General Hospital, 11527 Athens, Greece
| | - Vasileios Papastamopoulos
- 5th Department of Internal Medicine, Infectious Diseases Unit, Evaggelismos General Hospital, 10676 Athens, Greece
| | - Marika Kotsianopoulou
- Department of Public Health Policy, School of Public Health, University of West Attica, 11521 Athens, Greece
| |
Collapse
|
18
|
Abdoli A, Ghaffarifar F, Sharifi Z, Taghipour A. Toxoplasma gondii infection and testosterone alteration: A systematic review and meta-analyses. PLoS One 2024; 19:e0297362. [PMID: 38568993 PMCID: PMC10990213 DOI: 10.1371/journal.pone.0297362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 01/03/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Toxoplasma gondii (T. gondii) is a worldwide distributed protozoan parasite which has infected a wide range of warm-blooded animals and humans. The most common form of T. gondii infection is asymptomatic (latent); nevertheless, latent toxoplasmosis can induce various alterations of sex hormones, especially testosterone, in infected humans and animals. On the other hand, testosterone is involved in behavioral traits and reproductive functions in both sexes. Hence, the purpose of this systematic review is to summarize the available evidence regarding the association between T. gondii infection and testosterone alteration. METHODS In the setting of a systematic review, an electronic search (any date to 10 January 2023) without language restrictions was performed using Science Direct, Web of Science, PubMed, Scopus, and Google Scholar. The PRISMA guidelines were followed. Following the initial search, a total of 12,306 titles and abstracts were screened initially; 12,281 were excluded due to the lack of eligibility criteria or duplication. Finally, 24 articles met the included criteria. A mean±standard deviation (SD) was calculated to assess the difference of testosterone between T. gondii positive and T. gondii negative humans. The possibility of publication bias was assessed using Egger's regression. P-value < 0.05 was considered statistically significant. RESULTS This systematic review identified 24 articles (18 studies in humans and six studies in animals). Most human studies (13 out of 19) reported an increased level of testosterone following latent toxoplasmosis in males, while three studies reported decreased levels and two studies reported an insignificant change. Eleven articles (seven datasets in males and seven datasets in females) were eligible to be included in the data synthesis. Based on the random-effects model, the pooled mean± SD of testosterone in T. gondii positive than T. gondii negative was increased by 0.73 and 0.55 units in males and females, respectively. The Egger's regression did not detect a statistically significant publication bias in males and females (p = value = 0.95 and 0.71), respectively. Three studies in male animals (rats, mice, and spotted hyenas) and two studies in female animals (mice and spotted hyenas) reported a decline in testosterone in infected compared with non-infected animals. While, one study in female rats reported no significant changes of testosterone in infected than non-infected animals. Moreover, two studies in male rats reported an increased level of testosterone in infected than non-infected animals. CONCLUSIONS This study provides new insights about the association between T. gondii infection and testosterone alteration and identifies relevant data gaps that can inform and encourage further studies. The consequence of increased testosterone levels following T. gondii infection could partly be associated with increased sexual behavior and sexual transmission of the parasite. On the other hand, declining testosterone levels following T. gondii infection may be associated with male reproductive impairments, which were observed in T. gondii-infected humans and animals. Furthermore, these findings suggest the great need for more epidemiological and experimental investigations in depth to understand the relationship between T. gondii infection and testosterone alteration alongside with future consequences of testosterone alteration.
Collapse
Affiliation(s)
- Amir Abdoli
- Zoonoses Research Center, Jahrom University of Medical Sciences, Jahrom, Iran
- Department of Parasitology and Mycology, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Fatemeh Ghaffarifar
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zohreh Sharifi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Ali Taghipour
- Zoonoses Research Center, Jahrom University of Medical Sciences, Jahrom, Iran
- Department of Parasitology and Mycology, Jahrom University of Medical Sciences, Jahrom, Iran
| |
Collapse
|
19
|
Rahman SU, Weng TN, Qadeer A, Nawaz S, Ullah H, Chen CC. Omega-3 and omega-6 polyunsaturated fatty acids and their potential therapeutic role in protozoan infections. Front Immunol 2024; 15:1339470. [PMID: 38633251 PMCID: PMC11022163 DOI: 10.3389/fimmu.2024.1339470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/21/2024] [Indexed: 04/19/2024] Open
Abstract
Protozoa exert a serious global threat of growing concern to human, and animal, and there is a need for the advancement of novel therapeutic strategies to effectively treat or mitigate the impact of associated diseases. Omega polyunsaturated fatty acids (ω-PUFAs), including Omega-3 (ω-3) and omega-6 (ω-6), are constituents derived from various natural sources, have gained significant attention for their therapeutic role in parasitic infections and a variety of essential structural and regulatory functions in animals and humans. Both ω-3 and ω-6 decrease the growth and survival rate of parasites through metabolized anti-inflammatory mediators, such as lipoxins, resolvins, and protectins, and have both in vivo and in vitro protective effects against various protozoan infections. The ω-PUFAs have been shown to modulate the host immune response by a commonly known mechanism such as (inhibition of arachidonic acid (AA) metabolic process, production of anti-inflammatory mediators, modification of intracellular lipids, and activation of the nuclear receptor), and promotion of a shift towards a more effective immune defense against parasitic invaders by regulation the inflammation like prostaglandins, leukotrienes, thromboxane, are involved in controlling the inflammatory reaction. The immune modulation may involve reducing inflammation, enhancing phagocytosis, and suppressing parasitic virulence factors. The unique properties of ω-PUFAs could prevent protozoan infections, representing an important area of study. This review explores the clinical impact of ω-PUFAs against some protozoan infections, elucidating possible mechanisms of action and supportive therapy for preventing various parasitic infections in humans and animals, such as toxoplasmosis, malaria, coccidiosis, and chagas disease. ω-PUFAs show promise as a therapeutic approach for parasitic infections due to their direct anti-parasitic effects and their ability to modulate the host immune response. Additionally, we discuss current treatment options and suggest perspectives for future studies. This could potentially provide an alternative or supplementary treatment option for these complex global health problems.
Collapse
Affiliation(s)
- Sajid Ur Rahman
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Tzu-Nin Weng
- Department of Stomatology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Abdul Qadeer
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Saqib Nawaz
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Hanif Ullah
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- West China Hospital, School of Nursing, Sichuan University, Chengdu, China
| | - Chien-Chin Chen
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
- Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
- Doctoral Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
20
|
Andreou D, Steen NE, Mørch-Johnsen L, Jørgensen KN, Wortinger LA, Barth C, Szabo A, O'Connell KS, Lekva T, Hjell G, Johansen IT, Ormerod MBEG, Haukvik UK, Aukrust P, Djurovic S, Yolken RH, Andreassen OA, Ueland T, Agartz I. Toxoplasma gondii infection associated with inflammasome activation and neuronal injury. Sci Rep 2024; 14:5327. [PMID: 38438515 PMCID: PMC10912117 DOI: 10.1038/s41598-024-55887-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 02/28/2024] [Indexed: 03/06/2024] Open
Abstract
Toxoplasma gondii (TOXO) infection typically results in chronic latency due to its ability to form cysts in the brain and other organs. Latent toxoplasmosis could promote innate immune responses and impact brain function. A large body of evidence has linked TOXO infection to severe mental illness (SMI). We hypothesized that TOXO immunoglobulin G (IgG) seropositivity, reflecting previous infection and current latency, is associated with increased circulating neuron-specific enolase (NSE), a marker of brain damage, and interleukin-18 (IL-18), an innate immune marker, mainly in SMI. We included 735 patients with SMI (schizophrenia or bipolar spectrum) (mean age 32 years, 47% women), and 518 healthy controls (HC) (mean age 33 years, 43% women). TOXO IgG, expressed as seropositivity/seronegativity, NSE and IL-18 were measured with immunoassays. We searched for main and interaction effects of TOXO, patient/control status and sex on NSE and IL-18. In the whole sample as well as among patients and HC separately, IL-18 and NSE concentrations were positively correlated (p < 0.001). TOXO seropositive participants had significantly higher NSE (3713 vs. 2200 pg/ml, p < 0.001) and IL-18 levels (1068 vs. 674 pg/ml, p < 0.001) than seronegative participants, and evaluation within patients and HC separately showed similar results. Post-hoc analysis on cytomegalovirus and herpes simplex virus 1 IgG status showed no associations with NSE or IL-18 which may suggest TOXO specificity. These results may indicate ongoing inflammasome activation and neuronal injury in people with TOXO infections unrelated to diagnosis.
Collapse
Affiliation(s)
- Dimitrios Andreou
- Department of Psychiatric Research, Diakonhjemmet Hospital, Forskningsveien 7, 0373, Oslo, Norway.
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden.
| | - Nils Eiel Steen
- Department of Psychiatric Research, Diakonhjemmet Hospital, Forskningsveien 7, 0373, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Lynn Mørch-Johnsen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry and Department of Clinical Research, Østfold Hospital, Grålum, Norway
| | - Kjetil Nordbø Jørgensen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Vestre Viken Hospital Trust, Drammen, Norway
| | - Laura A Wortinger
- Department of Psychiatric Research, Diakonhjemmet Hospital, Forskningsveien 7, 0373, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Claudia Barth
- Department of Psychiatric Research, Diakonhjemmet Hospital, Forskningsveien 7, 0373, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Attila Szabo
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Kevin S O'Connell
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Gabriela Hjell
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry and Department of Clinical Research, Østfold Hospital, Grålum, Norway
| | - Ingrid Torp Johansen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Monica B E G Ormerod
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Unn K Haukvik
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Forensic Research and Education, Oslo University Hospital, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Ingrid Agartz
- Department of Psychiatric Research, Diakonhjemmet Hospital, Forskningsveien 7, 0373, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
21
|
Almurshidi BH, Fahmy Z, El-Shennawy A, Selim EAH, Hammam OA, Okasha H, Al-Hajj W, Mahmoud SA, Abuelenain GL. A multimodality therapeutic application on Toxoplasma gondii encephalitis utilizing Spiramycin and 'de novo' Ferula asafetida in immunodeficient mice. Parasite Immunol 2023; 45:e13014. [PMID: 37807942 DOI: 10.1111/pim.13014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/23/2023] [Accepted: 09/27/2023] [Indexed: 10/10/2023]
Abstract
This study investigated a 'de Novo' medicinal herb, Ferula asafetida (FA), against toxoplasma encephalitis either alone or combined with spiramycin (SP). Female Swiss-Webster mice (n = 72) were divided into three batches. Batch-I received no DMS to serve as an immunocompetent control, batch-II was immune-suppressed with the DMS (0.25 mg/g/day) for 14 days pre-infection, whilst batch-III was immune-suppressed with the DMS on the same day of infection. All experimental mice were inoculated with Toxoplasma gondii ME49 cysts (n = 75). Each batch was split into four subgroups: Mono-SP, mono-FA, combined drug (SP + FA), or neither. Therapies were administered on day zero of infection in batches (I and II) and 35 days post-infection in batch (III). Treatments lasted for 14 days, and mice were sacrificed 60 days post-infection. Histopathological changes, cysts load, and CD4 and CD8 T-cells were counted in brain tissues. The cyst-load count in mice receiving SP + FA was significantly (p < .0001) the least compared to the mono treatments in all protocols. Interestingly, the combined therapy demolished the T-cell subsets to zero in immunocompetent and immunocompromised infected mice. In conclusion, F. asafetida might be a powerfully natural, safe vehicle of SP in the digestive system and/or across the brain-blood barrier to control toxoplasmosis even through immunodeficient conditions.
Collapse
Affiliation(s)
| | - Zeinab Fahmy
- Immunology and Therapeutic Evaluation Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Amal El-Shennawy
- Immunology and Therapeutic Evaluation Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Eman A H Selim
- Immunology and Therapeutic Evaluation Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Olfat Ali Hammam
- Pathology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Hend Okasha
- Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | | | - Salma Awad Mahmoud
- Fatima College of Health Sciences, IAT, Abu Dhabi, UAE
- Cancer Science Institute, National University of Singapore, Singapore, Singapore
| | - Gehan Labib Abuelenain
- Immunology and Therapeutic Evaluation Department, Theodor Bilharz Research Institute, Giza, Egypt
| |
Collapse
|
22
|
Zarringhalam K, Ye S, Lou J, Rezvani Y, Gubbels MJ. Cell cycle-regulated ApiAP2s and parasite development: the Toxoplasma paradigm. Curr Opin Microbiol 2023; 76:102383. [PMID: 37898053 PMCID: PMC10840917 DOI: 10.1016/j.mib.2023.102383] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 08/11/2023] [Accepted: 08/27/2023] [Indexed: 10/30/2023]
Abstract
The cell division cycle of T. gondii is driven by cyclically expressed ApiAP2 transcription factors (AP2s) that promote gene sets (regulons) associated with specific biological functions. AP2s drive other AP2s, thereby propelling the progressive gene expression waves defining the lytic cycle. AP2s can act as dimers by themselves, in combination with other AP2s (constitutive or cyclical) or in complexes with epigenetic factors. Exit from the cell cycle into either the extracellular state or differentiation into bradyzoites results in major changes in gene expression. Surprisingly, both transitions lead to expression of a shared set of unique AP2s that suggest a shared stress response that, governed by the specific conditions, leads to different outcomes.
Collapse
Affiliation(s)
- Kourosh Zarringhalam
- Department of Mathematics, University of Massachusetts Boston, Boston, MA, USA; Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, USA.
| | - Sida Ye
- Department of Mathematics, University of Massachusetts Boston, Boston, MA, USA; Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, USA
| | - Jingjing Lou
- Department of Biology, Boston College, Chestnut Hill, MA, USA
| | - Yasaman Rezvani
- Department of Mathematics, University of Massachusetts Boston, Boston, MA, USA; Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, USA
| | | |
Collapse
|
23
|
Thind AC, Mota CM, Gonçalves APN, Sha J, Wohlschlegel JA, Mineo TWP, Bradley PJ. The Toxoplasma gondii effector GRA83 modulates the host's innate immune response to regulate parasite infection. mSphere 2023; 8:e0026323. [PMID: 37768053 PMCID: PMC10597413 DOI: 10.1128/msphere.00263-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/07/2023] [Indexed: 09/29/2023] Open
Abstract
Toxoplasma gondii's propensity to infect its host and cause disease is highly dependent on its ability to modulate host cell functions. One of the strategies the parasite uses to accomplish this is via the export of effector proteins from the secretory dense granules. Dense granule (GRA) proteins are known to play roles in nutrient acquisition, host cell cycle manipulation, and immune regulation. Here, we characterize a novel dense granule protein named GRA83, which localizes to the parasitophorous vacuole (PV) in tachyzoites and bradyzoites. Disruption of GRA83 results in increased virulence, weight loss, and parasitemia during the acute infection, as well as a marked increase in the cyst burden during the chronic infection. This increased parasitemia was associated with an accumulation of inflammatory infiltrates in tissues in both acute and chronic infections. Murine macrophages infected with ∆gra83 tachyzoites produced less interleukin-12 (IL-12) in vitro, which was confirmed with reduced IL-12 and interferon-gamma in vivo. This dysregulation of cytokines correlates with reduced nuclear translocation of the p65 subunit of the nuclear factor-κB (NF-κB) complex. While GRA15 similarly regulates NF-κB, infection with ∆gra83/∆gra15 parasites did not further reduce p65 translocation to the host cell nucleus, suggesting these GRAs function in converging pathways. We also used proximity labeling experiments to reveal candidate GRA83 interacting T. gondii-derived partners. Taken together, this work reveals a novel effector that stimulates the innate immune response, enabling the host to limit the parasite burden. Importance Toxoplasma gondii poses a significant public health concern as it is recognized as one of the leading foodborne pathogens in the United States. Infection with the parasite can cause congenital defects in neonates, life-threatening complications in immunosuppressed patients, and ocular disease. Specialized secretory organelles, including the dense granules, play an important role in the parasite's ability to efficiently invade and regulate components of the host's infection response machinery to limit parasite clearance and establish an acute infection. Toxoplasma's ability to avoid early clearance, while also successfully infecting the host long enough to establish a persistent chronic infection, is crucial in allowing for its transmission to a new host. While multiple GRAs directly modulate host signaling pathways, they do so in various ways highlighting the parasite's diverse arsenal of effectors that govern infection. Understanding how parasite-derived effectors harness host functions to evade defenses yet ensure a robust infection is important for understanding the complexity of the pathogen's tightly regulated infection. In this study, we characterize a novel secreted protein named GRA83 that stimulates the host cell's response to limit infection.
Collapse
Affiliation(s)
- Amara C. Thind
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Caroline M. Mota
- Laboratory of Immunoparasitology “Dr. Mário Endsfeldz Camargo,” Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Ana Paula N. Gonçalves
- Laboratory of Immunoparasitology “Dr. Mário Endsfeldz Camargo,” Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Jihui Sha
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - James A. Wohlschlegel
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Tiago W. P. Mineo
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
- Laboratory of Immunoparasitology “Dr. Mário Endsfeldz Camargo,” Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Peter J. Bradley
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
24
|
Holmes MJ, Bastos MS, Dey V, Severo V, Wek RC, Sullivan WJ. mRNA cap-binding protein eIF4E1 is a novel regulator of Toxoplasma gondii latency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.09.561274. [PMID: 37873335 PMCID: PMC10592687 DOI: 10.1101/2023.10.09.561274] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The protozoan parasite Toxoplasma gondii causes serious opportunistic disease due to its ability to persist in patients as latent tissue cysts. The molecular mechanisms coordinating conversion between proliferative parasites (tachyzoites) and dormant cysts (bradyzoites) are not fully understood. We previously showed that phosphorylation of eIF2α accompanies bradyzoite formation, suggesting that this clinically relevant process involves regulation of mRNA translation. In this study, we investigated the composition and role of eIF4F multi-subunit complexes in translational control. Using CLIPseq, we find that the cap-binding subunit, eIF4E1, localizes to the 5'-end of all tachyzoite mRNAs, many of which show evidence of stemming from heterogenous transcriptional start sites. We further show that eIF4E1 operates as the predominant cap-binding protein in two distinct eIF4F complexes. Using genetic and pharmacological approaches, we found that eIF4E1 deficiency triggers efficient spontaneous formation of bradyzoites without stress induction. Consistent with this result, we also show that stress-induced bradyzoites exhibit reduced eIF4E1 expression. Overall, our findings establish a novel role for eIF4F in translational control required for parasite latency and microbial persistence.
Collapse
Affiliation(s)
- Michael J. Holmes
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis IN
| | - Matheus S. Bastos
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis IN
| | - Vishakha Dey
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis IN
| | - Vanessa Severo
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis IN
| | - Ronald C. Wek
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis IN
| | - William J. Sullivan
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis IN
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis IN
| |
Collapse
|
25
|
Doghish AS, Ali MA, Elrebehy MA, Mohamed HH, Mansour R, Ghanem A, Hassan A, Elballal MS, Elazazy O, Elesawy AE, Abdel Mageed SS, Nassar YA, Mohammed OA, Abulsoud AI. The interplay between toxoplasmosis and host miRNAs: Mechanisms and consequences. Pathol Res Pract 2023; 250:154790. [PMID: 37683390 DOI: 10.1016/j.prp.2023.154790] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023]
Abstract
Toxoplasmosis is one of the highly prevalent zoonotic diseases worldwide caused by the parasite Toxoplasma gondii (T. gondii). The infection with T. gondii could pass unidentified in immunocompetent individuals; however, latent cysts remain dormant in their digestive tract, but they could be shed and excreted with feces infesting the environment. However, active toxoplasmosis can create serious consequences, particularly in newborns and infected persons with compromised immunity. These complications include ocular toxoplasmosis, in which most cases cannot be treated. Additionally, it caused many stillbirths and miscarriages. Circulating miRNAs are important regulatory molecules ensuring that the normal physiological role of various organs is harmonious. Upon infection with T. gondii, the tightly regulated miRNA profile is disrupted to favor the parasite's survival and further participate in the disease pathogenesis. Interestingly, this dysregulated profile could be useful in acute and chronic disease discrimination and in providing insights into the pathomechanisms of the disease. Thus, this review sheds light on the various roles of miRNAs in signaling pathways regulation involved in the pathogenesis of T. gondii and provides insights into the application of miRNAs clinically for its diagnosis and prognosis.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt.
| | - Mohamed A Ali
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt.
| | - Hend H Mohamed
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Reda Mansour
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan 11795, Egypt; Biology Department, School of Biotechnology, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Aml Ghanem
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed Hassan
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan 11795, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ola Elazazy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Yara A Nassar
- Biology Department, School of Biotechnology, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Osama A Mohammed
- Department of Clinical Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt; Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| |
Collapse
|
26
|
Tarannum A, Rodríguez-Almonacid CC, Salazar-Bravo J, Karamysheva ZN. Molecular Mechanisms of Persistence in Protozoan Parasites. Microorganisms 2023; 11:2248. [PMID: 37764092 PMCID: PMC10534552 DOI: 10.3390/microorganisms11092248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Protozoan parasites are known for their remarkable capacity to persist within the bodies of vertebrate hosts, which frequently results in prolonged infections and the recurrence of diseases. Understanding the molecular mechanisms that underlie the event of persistence is of paramount significance to develop innovative therapeutic approaches, given that these pathways still need to be thoroughly elucidated. The present article provides a comprehensive overview of the latest developments in the investigation of protozoan persistence in vertebrate hosts. The focus is primarily on the function of persisters, their formation within the host, and the specific molecular interactions between host and parasite while they persist. Additionally, we examine the metabolomic, transcriptional, and translational changes that protozoan parasites undergo during persistence within vertebrate hosts, focusing on major parasites such as Plasmodium spp., Trypanosoma spp., Leishmania spp., and Toxoplasma spp. Key findings of our study suggest that protozoan parasites deploy several molecular and physiological strategies to evade the host immune surveillance and sustain their persistence. Furthermore, some parasites undergo stage differentiation, enabling them to acclimate to varying host environments and immune challenges. More often, stressors such as drug exposure were demonstrated to impact the formation of protozoan persisters significantly. Understanding the molecular mechanisms regulating the persistence of protozoan parasites in vertebrate hosts can reinvigorate our current insights into host-parasite interactions and facilitate the development of more efficacious disease therapeutics.
Collapse
Affiliation(s)
| | | | | | - Zemfira N. Karamysheva
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA; (A.T.); (C.C.R.-A.); (J.S.-B.)
| |
Collapse
|
27
|
Fahmy MEA, Shalaby MA, Issa R, Badawi M, Magdy M, Afife AA, Abdel-Aal AA. Ivermectin modulated cerebral γ-aminobutyric acid (GABA) and reduced the number of chronic Toxoplasma gondii cysts significantly in the brains of immunocompromised mice. J Parasit Dis 2023; 47:635-643. [PMID: 37520203 PMCID: PMC10382416 DOI: 10.1007/s12639-023-01608-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/09/2023] [Indexed: 08/01/2023] Open
Abstract
Disruption of GABAergic signaling could exaggerate the inflammatory reaction associated with Toxoplasma gondii infection, as well as produce neurophysiological consequences including seizures that occur within the brain tissues. The current study aimed to evaluate the efficacy of ivermectin (IVM) in treating latent cerebral toxoplasmosis and define its role in the neuromodulation of cerebral tissue GABA expression, conducted in an immunocompromised dexamethasone-treated mouse model infected with the ME49 Toxoplasma strain. The control (non-infected non-treated) group showed a mean of 22.1 ± 0.71 for local expression of GABA. Significantly lower expression (3.78 ± 1.38) was recorded in the infected non-treated group (p ≤ 0.05). On the contrary, a significantly higher expression was reported in the group infected and treated with IVM than in the infected non-treated group (19.8 ± 0.8). While the infected spiramycin (SP)-treated group reported a significantly lower level than the control. Non-infected groups that received only IVM or SP recorded 22.3 ± 0.45 and 22 ± 0.89 respectively with no significant difference. IVM is shown in this work, not only to reduce the size and the number of Toxoplasma cystic lesions within the brain significantly with a reduction rate of 68.85% but to also increase the level of GABA local expression significantly in addition to improving cerebral histopathology. Thus, IVM by its ability to modulate GABA expression may improve such clinical situations, if used as a treatment either exclusively or in combination with other medications.
Collapse
Affiliation(s)
| | - Maisa Ahmed Shalaby
- Medical Parasitology Department, Theodor Bilharz Research Institute (TBRI), Giza, Egypt
| | - Ragaa Issa
- Departement of Parasitology, Research Institute of Ophthalmology, Giza, Egypt
| | - Manal Badawi
- Departement of Pathology, National Research Centre, Giza, Egypt
| | - Mona Magdy
- Department of Pathology, Theodor Bilharz Research Institute (TBRI), Giza, Egypt
| | - Adam Ashraf Afife
- College of Life Sciences, Faculty of Medicine, Leicester University, Leicester, UK
| | - Amany Ahmed Abdel-Aal
- Department of Medical Parasitology, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Postgraduate Studies and Scientific Research, Armed Forces College of Medicine (AFCM), Cairo, Egypt
| |
Collapse
|
28
|
Hildebrandt F, Mohammed M, Dziedziech A, Bhandage AK, Divne AM, Barrenäs F, Barragan A, Henriksson J, Ankarklev J. scDual-Seq of Toxoplasma gondii-infected mouse BMDCs reveals heterogeneity and differential infection dynamics. Front Immunol 2023; 14:1224591. [PMID: 37575232 PMCID: PMC10415529 DOI: 10.3389/fimmu.2023.1224591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/06/2023] [Indexed: 08/15/2023] Open
Abstract
Dendritic cells and macrophages are integral parts of the innate immune system and gatekeepers against infection. The protozoan pathogen, Toxoplasma gondii, is known to hijack host immune cells and modulate their immune response, making it a compelling model to study host-pathogen interactions. Here we utilize single cell Dual RNA-seq to parse out heterogeneous transcription of mouse bone marrow-derived dendritic cells (BMDCs) infected with two distinct genotypes of T. gondii parasites, over multiple time points post infection. We show that the BMDCs elicit differential responses towards T. gondii infection and that the two parasite lineages distinctly manipulate subpopulations of infected BMDCs. Co-expression networks define host and parasite genes, with implications for modulation of host immunity. Integrative analysis validates previously established immune pathways and additionally, suggests novel candidate genes involved in host-pathogen interactions. Altogether, this study provides a comprehensive resource for characterizing host-pathogen interplay at high-resolution.
Collapse
Affiliation(s)
- Franziska Hildebrandt
- Department of Molecular Biosciences, The Wenner Gren Institute, Stockholm University, Stockholm, Sweden
| | - Mubasher Mohammed
- Department of Molecular Biosciences, The Wenner Gren Institute, Stockholm University, Stockholm, Sweden
| | - Alexis Dziedziech
- Department of Molecular Biosciences, The Wenner Gren Institute, Stockholm University, Stockholm, Sweden
- Department of Global Health, Institut Pasteur, Paris, France
| | - Amol K. Bhandage
- Department of Molecular Biosciences, The Wenner Gren Institute, Stockholm University, Stockholm, Sweden
| | - Anna-Maria Divne
- Microbial Single Cell Genomics Facility, SciLifeLab, Biomedical Center (BMC) Uppsala University, Uppsala, Sweden
| | - Fredrik Barrenäs
- Department of Molecular Biosciences, The Wenner Gren Institute, Stockholm University, Stockholm, Sweden
| | - Antonio Barragan
- Department of Molecular Biosciences, The Wenner Gren Institute, Stockholm University, Stockholm, Sweden
| | - Johan Henriksson
- Laboratory of Molecular Infection Medicine Sweden (MIMS), Umeå Center for Microbial Research, Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Johan Ankarklev
- Department of Molecular Biosciences, The Wenner Gren Institute, Stockholm University, Stockholm, Sweden
- Microbial Single Cell Genomics Facility, SciLifeLab, Biomedical Center (BMC) Uppsala University, Uppsala, Sweden
| |
Collapse
|
29
|
Thind AC, Mota CM, Gonçalves APN, Sha J, Wohlschlegel JA, Mineo TWP, Bradley PJ. The Toxoplasma gondii effector GRA83 modulates the host's innate immune response to regulate parasite infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.31.543158. [PMID: 37398161 PMCID: PMC10312501 DOI: 10.1101/2023.05.31.543158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Toxoplasma gondii 's propensity to infect its host and cause disease is highly dependent on its ability to modulate host cell functions. One of the strategies the parasite uses to accomplish this is via the export of effector proteins from the secretory dense granules. Dense granule (GRA) proteins are known to play roles in nutrient acquisition, host cell cycle manipulation, and immune regulation. Here, we characterize a novel dense granule protein named GRA83, which localizes to the parasitophorous vacuole in tachyzoites and bradyzoites. Disruption of GRA83 results in increased virulence, weight loss, and parasitemia during the acute infection, as well as a marked increase in the cyst burden during the chronic infection. This increased parasitemia was associated with an accumulation of inflammatory infiltrates in tissues in both the acute and chronic infection. Murine macrophages infected with Δ gra83 tachyzoites produced less interleukin-12 (IL-12) in vitro , which was confirmed with reduced IL-12 and interferon gamma (IFN-γ) in vivo . This dysregulation of cytokines correlates with reduced nuclear translocation of the p65 subunit of the NF-κB complex. While GRA15 similarly regulates NF-κB, infection with Δ gra83/ Δ gra15 parasites did not further reduce p65 translocation to the host cell nucleus, suggesting these GRAs function in converging pathways. We also used proximity labelling experiments to reveal candidate GRA83 interacting T. gondii derived partners. Taken together, this work reveals a novel effector that stimulates the innate immune response, enabling the host to limit parasite burden. Importance Toxoplasma gondii poses a significant public health concern as it is recognized as one of the leading foodborne pathogens in the United States. Infection with the parasite can cause congenital defects in neonates, life-threatening complications in immunosuppressed patients, and ocular disease. Specialized secretory organelles, including the dense granules, play an important role in the parasite's ability to efficiently invade and regulate components of the host's infection response machinery to limit parasite clearance and establish an acute infection. Toxoplasma' s ability to avoid early clearance, while also successfully infecting the host long enough to establish a persistent chronic infection, is crucial in allowing for its transmission to a new host. While multiple GRAs directly modulate host signaling pathways, they do so in various ways highlighting the parasite's diverse arsenal of effectors that govern infection. Understanding how parasite-derived effectors harness host functions to evade defenses yet ensure a robust infection are important for understanding the complexity of the pathogen's tightly regulated infection. In this study, we characterize a novel secreted protein named GRA83 that stimulates the host cell's response to limit infection.
Collapse
|
30
|
Sena F, Cancela S, Bollati-Fogolín M, Pagotto R, Francia ME. Exploring Toxoplasma gondii´s Biology within the Intestinal Epithelium: intestinal-derived models to unravel sexual differentiation. Front Cell Infect Microbiol 2023; 13:1134471. [PMID: 37313339 PMCID: PMC10258352 DOI: 10.3389/fcimb.2023.1134471] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/25/2023] [Indexed: 06/15/2023] Open
Abstract
A variety of intestinal-derived culture systems have been developed to mimic in vivo cell behavior and organization, incorporating different tissue and microenvironmental elements. Great insight into the biology of the causative agent of toxoplasmosis, Toxoplasma gondii, has been attained by using diverse in vitro cellular models. Nonetheless, there are still processes key to its transmission and persistence which remain to be elucidated, such as the mechanisms underlying its systemic dissemination and sexual differentiation both of which occur at the intestinal level. Because this event occurs in a complex and specific cellular environment (the intestine upon ingestion of infective forms, and the feline intestine, respectively), traditional reductionist in vitro cellular models fail to recreate conditions resembling in vivo physiology. The development of new biomaterials and the advances in cell culture knowledge have opened the door to a next generation of more physiologically relevant cellular models. Among them, organoids have become a valuable tool for unmasking the underlying mechanism involved in T. gondii sexual differentiation. Murine-derived intestinal organoids mimicking the biochemistry of the feline intestine have allowed the generation of pre-sexual and sexual stages of T. gondii for the first time in vitro, opening a window of opportunity to tackling these stages by "felinizing" a wide variety of animal cell cultures. Here, we reviewed intestinal in vitro and ex vivo models and discussed their strengths and limitations in the context of a quest for faithful models to in vitro emulate the biology of the enteric stages of T. gondii.
Collapse
Affiliation(s)
- Florencia Sena
- Laboratory of Apicomplexan Biology, Institut Pasteur Montevideo, Montevideo, Uruguay
- Laboratorio de Bioquímica, Departamento de Biología Vegetal, Universidad de la República, Montevideo, Uruguay
| | - Saira Cancela
- Cell Biology Unit, Institut Pasteur Montevideo, Montevideo, Uruguay
- Molecular, Cellular, and Animal Technology Program (ProTeMCA), Institut Pasteur Montevideo, Montevideo, Uruguay
| | - Mariela Bollati-Fogolín
- Cell Biology Unit, Institut Pasteur Montevideo, Montevideo, Uruguay
- Molecular, Cellular, and Animal Technology Program (ProTeMCA), Institut Pasteur Montevideo, Montevideo, Uruguay
| | - Romina Pagotto
- Cell Biology Unit, Institut Pasteur Montevideo, Montevideo, Uruguay
| | - María E. Francia
- Laboratory of Apicomplexan Biology, Institut Pasteur Montevideo, Montevideo, Uruguay
- Departamento de Parasitología y Micología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
31
|
Kochanowsky JA, Chandrasekaran S, Sanchez JR, Thomas KK, Koshy AA. ROP16-mediated activation of STAT6 enhances cyst development of type III Toxoplasma gondii in neurons. PLoS Pathog 2023; 19:e1011347. [PMID: 37068104 PMCID: PMC10138205 DOI: 10.1371/journal.ppat.1011347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 04/27/2023] [Accepted: 04/07/2023] [Indexed: 04/18/2023] Open
Abstract
Toxoplasma gondii establishes a long-lived latent infection in the central nervous system (CNS) of its hosts. Reactivation in immunocompromised individuals can lead to life threatening disease. Latent infection is driven by the ability of the parasite to convert from the acute-stage tachyzoite to the latent-stage bradyzoite which resides in long-lived intracellular cysts. While much work has focused on the parasitic factors that drive cyst development, the host factors that influence encystment are not well defined. Here we show that a polymorphic secreted parasite kinase (ROP16), that phosphorylates host cell proteins, mediates efficient encystment of T. gondii in a stress-induced model of encystment and primary neuronal cell cultures (PNCs) in a strain-specific manner. Using short-hairpin RNA (shRNA) knockdowns in human foreskin fibroblasts (HFFs) and PNCs from transgenic mice, we determined that ROP16's cyst enhancing abilities are mediated, in part, by phosphorylation-and therefore activation-of the host cell transcription factor STAT6. To test the role of STAT6 in vivo, we infected wild-type (WT) and STAT6KO mice, finding that, compared to WT mice, STAT6KO mice have a decrease in CNS cyst burden but not overall parasite burden or dissemination to the CNS. Finally, we found a similar ROP16-dependent encystment defect in human pluripotent stem cell-derived neurons. Together, these findings identify a host cell factor (STAT6) that T. gondii manipulates in a strain-specific manner to generate a favorable encystment environment.
Collapse
Affiliation(s)
- Joshua A. Kochanowsky
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | | | - Jacqueline R. Sanchez
- Postbaccalaureate Research Education Program, University of Arizona, Tucson, Arizona, United States of America
| | - Kaitlin K. Thomas
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Anita A. Koshy
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Department of Neurology, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
32
|
Recombinant Mouse Prolactin Confers Partial Protection Against Toxoplasma gondii Infection in a Pre-treated Experimental Murine Model. Acta Parasitol 2023; 68:182-193. [PMID: 36542295 DOI: 10.1007/s11686-022-00651-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Toxoplasmosis is one of the most important health-threatening diseases with worldwide distribution and global impact. It is caused by Toxoplasma gondii (T. gondii), an intracellular apicomplexan parasite that can evade the host immune responses and establish a chronic infection. The available treatments are not efficient against this stage and have many adverse effects. There are no available effective vaccines, apart from Toxovax®, which is used in sheep to prevent abortion. Studies documented that prolactin (PRL) had in vivo and in vitro anti-Toxoplasma effects. Detailed research was recommended about the mechanisms of such inhibitory effects. AIM This study was designed to assess the possible protective role of the recombinant prolactin (rPRL) against T. gondii. MATERIALS AND METHODS Sixty experimentally infected mice with T. gondii were used. The treated mice received rPRL for five days before infection. Serum prolactin levels were measured; survival rate was monitored; number, size, and DNA of T. gondii cysts in the brain were measured; and histopathological and immunological studies were done. RESULTS There was a significant increase in the survival rate of the rPRL-treated mice, a significant decrease in the number, size, and DNA amount of T. gondii cysts in the brain with a noticeable improvement of histopathological lesions in the brain and liver tissues when compared to the infected untreated group. These effects seem to be achieved through stimulating humoral and cell-mediated immune responses that were evident by the significant rise in serum levels of anti-Toxoplasma IgM, IFN-γ, and TNF-α. CONCLUSION The rPRL elicited robust immune responses, which provided efficient protection against murine T. gondii infection.
Collapse
|
33
|
Warschkau D, Seeber F. Advances towards the complete in vitro life cycle of Toxoplasma gondii. Fac Rev 2023; 12:1. [PMID: 36846606 PMCID: PMC9944905 DOI: 10.12703/r/12-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
The full life cycle of Toxoplasma gondii cannot be recapitulated in vitro, and access to certain stages, such as mature tissue cysts (bradyzoites) and oocysts (sporozoites), traditionally requires animal experiments. This has greatly hindered the study of the biology of these morphologically and metabolically distinct stages, which are essential for the infection of humans and animals. However, several breakthrough advances have been made in recent years towards obtaining these life stages in vitro, such as the discovery of several molecular factors that induce differentiation and commitment to the sexual cycle, and different culture methods that use, for example, myotubes and intestinal organoids to obtain mature bradyzoites and different sexual stages of the parasite. We review these novel tools and approaches, highlight their limitations and challenges, and discuss what research questions can already be answered with these models. We finally identify future routes for recapitulating the entire sexual cycle in vitro.
Collapse
Affiliation(s)
- David Warschkau
- FG16: Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institut, Berlin, Germany
| | - Frank Seeber
- FG16: Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institut, Berlin, Germany
| |
Collapse
|
34
|
Xiao J, Li Y, Rowley T, Huang J, Yolken RH, Viscidi RP. Immunotherapy targeting the PD-1 pathway alleviates neuroinflammation caused by chronic Toxoplasma infection. Sci Rep 2023; 13:1288. [PMID: 36690687 PMCID: PMC9870997 DOI: 10.1038/s41598-023-28322-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/17/2023] [Indexed: 01/25/2023] Open
Abstract
Toxoplasma gondii can infect the host brain and trigger neuroinflammation. Such neuroinflammation might persist for years if the infection is not resolved, resulting in harmful outcomes for the brain. We have previously demonstrated the efficacy of immunotherapy targeting the programmed cell death protein 1 (PD-1) pathway on clearance of Toxoplasma tissue cysts. We aimed to test whether parasite clearance would lead to the resolution of neuroinflammation in infected brains. We established chronic Toxoplasma infection in BALB/c mice using the cyst-forming Prugniaud strain. Mice then received αPD-L1 or isotype control antibodies. After completion of the therapy, mice were euthanized six weeks later. The number of brain tissue cysts, Toxoplasma-specific CD8 + T cell proliferation and IFN-γ secretion, serum cytokine and chemokine levels, and CNS inflammation were measured. In αPD-L1-treated mice, we observed reduced brain tissue cysts, increased spleen weight, elevated IFN-γ production by antigen-specific CD8 + T cells, and a general increase in multiple serum cytokines and chemokines. Importantly, αPD-L1-treated mice displayed attenuation of meningeal lymphocytes, reactive astrocytes, and C1q expression. The reduction in inflammation-related proteins is correlated with reduced parasite burden. These results suggest that promoting systemic immunity results in parasite clearance, which in turn alleviates neuroinflammation. Our study may have implications for some brain infections where neuroinflammation is a critical component.
Collapse
Affiliation(s)
- Jianchun Xiao
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA.
| | - Ye Li
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Treva Rowley
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Jing Huang
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Raphael P Viscidi
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| |
Collapse
|
35
|
Dámek F, Swart A, Waap H, Jokelainen P, Le Roux D, Deksne G, Deng H, Schares G, Lundén A, Álvarez-García G, Betson M, Davidson RK, Györke A, Antolová D, Hurníková Z, Wisselink HJ, Sroka J, van der Giessen JWB, Blaga R, Opsteegh M. Systematic Review and Modelling of Age-Dependent Prevalence of Toxoplasma gondii in Livestock, Wildlife and Felids in Europe. Pathogens 2023; 12:pathogens12010097. [PMID: 36678447 PMCID: PMC9865579 DOI: 10.3390/pathogens12010097] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Toxoplasma gondii is a zoonotic parasite of importance to both human and animal health. The parasite has various transmission routes, and the meat of infected animals appears to be a major source of human infections in Europe. We aimed to estimate T. gondii prevalence in a selection of animal host species. A systematic literature review resulting in 226 eligible publications was carried out, and serological data were analyzed using an age-dependent Bayesian hierarchical model to obtain estimates for the regional T. gondii seroprevalence in livestock, wildlife, and felids. Prevalence estimates varied between species, regions, indoor/outdoor rearing, and types of detection methods applied. The lowest estimated seroprevalence was observed for indoor-kept lagomorphs at 4.8% (95% CI: 1.8-7.5%) and the highest for outdoor-kept sheep at 63.3% (95% CI: 53.0-79.3%). Overall, T. gondii seroprevalence estimates were highest within Eastern Europe, whilst being lowest in Northern Europe. Prevalence data based on direct detection methods were scarce and were not modelled but rather directly summarized by species. The outcomes of the meta-analysis can be used to extrapolate data to areas with a lack of data and provide valuable inputs for future source attribution approaches aiming to estimate the relative contribution of different sources of T. gondii human infection.
Collapse
Affiliation(s)
- Filip Dámek
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, Laboratoire de Santé Animale, BIPAR, F-94700 Maisons-Alfort, France
| | - Arno Swart
- Centre for Infectious Disease Control—Zoonoses and Environmental Microbiology, National Institute for Public Health and the Environment, Antonie van Leeuwenhoeklaan 9, P.O. Box 1, 3720 BA Bilthoven, The Netherlands
| | - Helga Waap
- Laboratório de Parasitologia, Instituto Nacional de Investigação Agrária e Veterinária, 2780-157 Oeiras, Portugal
| | - Pikka Jokelainen
- Infectious Disease Preparedness, Statens Serum Institut, 2300 Copenhagen, Denmark
| | - Delphine Le Roux
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, Laboratoire de Santé Animale, BIPAR, F-94700 Maisons-Alfort, France
| | - Gunita Deksne
- Institute of Food Safety, Animal Health and Environment BIOR, LV-1076 Riga, Latvia
| | - Huifang Deng
- Centre for Infectious Disease Control—Zoonoses and Environmental Microbiology, National Institute for Public Health and the Environment, Antonie van Leeuwenhoeklaan 9, P.O. Box 1, 3720 BA Bilthoven, The Netherlands
| | - Gereon Schares
- Institute of Epidemiology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, 17493 Greifswald, Germany
| | - Anna Lundén
- Department of Microbiology, National Veterinary Institute, 75189 Uppsala, Sweden
| | - Gema Álvarez-García
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, 28040 Madrid, Spain
| | - Martha Betson
- School of Veterinary Medicine, University of Surrey, Guildford GU2 7AL, UK
| | - Rebecca K. Davidson
- Food Safety and Animal Health, Norwegian Veterinary Institute, 9016 Tromsø, Norway
| | - Adriana Györke
- Department of Parasitology and Parasitic Diseases, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 400372 Cluj-Napoca, Romania
| | - Daniela Antolová
- Institute of Parasitology, Slovak Academy of Sciences, 040 01 Košice, Slovakia
| | - Zuzana Hurníková
- Institute of Parasitology, Slovak Academy of Sciences, 040 01 Košice, Slovakia
| | - Henk J. Wisselink
- Wageningen Bioveterinary Research, Wageningen University & Research, P.O. Box 65, 8200 AB Lelystad, The Netherlands
| | - Jacek Sroka
- Department of Parasitology and Invasive Diseases, National Veterinary Research Institute, 24-100 Pulawy, Poland
| | - Joke W. B. van der Giessen
- Centre for Infectious Disease Control—Zoonoses and Environmental Microbiology, National Institute for Public Health and the Environment, Antonie van Leeuwenhoeklaan 9, P.O. Box 1, 3720 BA Bilthoven, The Netherlands
| | - Radu Blaga
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, Laboratoire de Santé Animale, BIPAR, F-94700 Maisons-Alfort, France
| | - Marieke Opsteegh
- Centre for Infectious Disease Control—Zoonoses and Environmental Microbiology, National Institute for Public Health and the Environment, Antonie van Leeuwenhoeklaan 9, P.O. Box 1, 3720 BA Bilthoven, The Netherlands
- Correspondence: ; Tel.: +31-6-29651388
| |
Collapse
|
36
|
Ifijen IH, Atoe B, Ekun RO, Ighodaro A, Odiachi IJ. Treatments of Mycobacterium tuberculosis and Toxoplasma gondii with Selenium Nanoparticles. BIONANOSCIENCE 2023; 13:249-277. [PMID: 36687337 PMCID: PMC9838309 DOI: 10.1007/s12668-023-01059-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2023] [Indexed: 01/13/2023]
Abstract
Toxoplasma gondii and Mycobacterium tuberculosis are pathogens that are harmful to humans. When these diseases interact in humans, the result is typically fatal to the public health. Several investigations on the relationship between M. tuberculosis and T. gondii infections have found that there is a strong correlation between them with each infection having a reciprocal effect on the other. TB may contribute to the reactivation of innate toxoplasmosis or enhance susceptibility to a new infection, and toxoplasma co-infection may worsen the severity of pulmonary tuberculosis. As a consequence, there is an earnest and urgent necessity to generate novel therapeutics that can subdue these challenges. Selenium nanostructures' compelling properties have been shown to be a successful treatment for Mycobacterium TB and Toxoplasma gondii. Despite the fact that selenium (Se) offers many health advantages for people, it also has a narrow therapeutic window; therefore, consuming too much of either inorganic or organic compounds based on selenium can be hazardous. Compared to both inorganic and organic Se, Se nanoparticles (SeNPs) are less hazardous. They are biocompatible and excellent in selectively targeting specific cells. As a consequence, this review conducted a summary of the efficacy of biogenic Se NPs in the treatment of tuberculosis (TB) and toxoplasmosis. Mycobacterium tuberculosis, Toxoplasma gondii, and their co-infection were all briefly described.
Collapse
Affiliation(s)
- Ikhazuagbe H. Ifijen
- Department of Research Outreach, Rubber Research Institute of Nigeria, Iyanomo, P.M.B, 1049, Benin City, Nigeria
| | - Best Atoe
- Department of Daily Need, Worldwide Healthcare, 100, Textile Mill Road, Benin City, Edo State Nigeria
| | - Raphael O. Ekun
- grid.440833.80000 0004 0642 9705Department of Electrical Electronics, Cyprus International University, Haspolat, Lefkosa, North Cyprus Mersin 10 Turkey
| | - Augustine Ighodaro
- Depatment of Aseptic Quality, Quantum Pharmaceuticals, Quantum House, Durham, UK
| | - Ifeanyi J. Odiachi
- grid.461933.a0000 0004 0446 5040Department of Science Laboratory Technology, Delta State Polytechnic Ogwashi-Uku, Ogwashi-Uku, Nigeria
| |
Collapse
|
37
|
Elmasry A, Aboukamar WA, Hany H, Elmehankar MS. The immunomodulatory effects of roflumilast on tachyzoite-bradyzoite transition in a murine model of Toxoplasma gondii. Int Immunopharmacol 2022; 113:109348. [DOI: 10.1016/j.intimp.2022.109348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022]
|
38
|
Keroack CD, Duraisingh MT. Molecular mechanisms of cellular quiescence in apicomplexan parasites. Curr Opin Microbiol 2022; 70:102223. [PMID: 36274498 DOI: 10.1016/j.mib.2022.102223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 01/25/2023]
Abstract
Quiescence is a reversible nonproliferative cellular state that allows organisms to persist through unfavorable conditions. Quiescence can be stimulated by a wide range of external or intrinsic factors. Cells undergo a coordinated molecular program to enter and exit from the quiescent state, which is governed by signaling, transcriptional and translational changes, epigenetic mechanisms, metabolic switches, and changes in cellular architecture. These mechanisms have been extensively studied in model organisms, and a growing number of studies have identified conserved mechanisms in apicomplexan parasites. Quiescence in the context of a parasitic infection has significant clinical impact: quiescent forms may underlie treatment failures, relapsing infections, and stress tolerance. Here, we review the latest understanding of quiescence in apicomplexa, synthesizing these studies to highlight conserved mechanisms, and identifying technologies to assist in further characterization of quiescence. Understanding conserved mechanisms of quiescence in apicomplexans will provide avenues for transmission prevention and radical cure of infections.
Collapse
|
39
|
Pan M, Ge CC, Fan YM, Jin QW, Shen B, Huang SY. The determinants regulating Toxoplasma gondii bradyzoite development. Front Microbiol 2022; 13:1027073. [PMID: 36439853 PMCID: PMC9691885 DOI: 10.3389/fmicb.2022.1027073] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/24/2022] [Indexed: 11/04/2023] Open
Abstract
Toxoplasma gondii is an obligate intracellular zoonotic pathogen capable of infecting almost all cells of warm-blooded vertebrates. In intermediate hosts, this parasite reproduces asexually in two forms, the tachyzoite form during acute infection that proliferates rapidly and the bradyzoite form during chronic infection that grows slowly. Depending on the growth condition, the two forms can interconvert. The conversion of tachyzoites to bradyzoites is critical for T. gondii transmission, and the reactivation of persistent bradyzoites in intermediate hosts may lead to symptomatic toxoplasmosis. However, the mechanisms that control bradyzoite differentiation have not been well studied. Here, we review recent advances in the study of bradyzoite biology and stage conversion, aiming to highlight the determinants associated with bradyzoite development and provide insights to design better strategies for controlling toxoplasmosis.
Collapse
Affiliation(s)
- Ming Pan
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Ceng-Ceng Ge
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yi-Min Fan
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Qi-Wang Jin
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Bang Shen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Si-Yang Huang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
40
|
A Signaling Factor Linked to Toxoplasma gondii Guanylate Cyclase Complex Controls Invasion and Egress during Acute and Chronic Infection. mBio 2022; 13:e0196522. [PMID: 36200777 PMCID: PMC9600588 DOI: 10.1128/mbio.01965-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Toxoplasma gondii is an intracellular apicomplexan parasite that relies on cyclic GMP (cGMP)-dependent signaling to trigger timely egress from host cells in response to extrinsic and intrinsic signals. A guanylate cyclase (GC) complex, conserved across the Apicomplexa, plays a pivotal role in integrating these signals, such as the key lipid mediator phosphatidic acid and changes in pH and ionic composition. This complex is composed of an atypical GC fused to a flippase-like P4-ATPase domain and assembled with the cell division control protein CDC50.1 and a unique GC organizer (UGO). While the dissemination of the fast-replicating tachyzoites responsible for acute infection is well understood, it is less clear if the cyst-forming bradyzoites can disseminate and contribute to cyst burden. Here, we characterized a novel component of the GC complex recently termed signaling linking factor (SLF). Tachyzoites conditionally depleted in SLF are impaired in microneme exocytosis, conoid extrusion, and motility and hence unable to invade and egress. A stage-specific promoter swap strategy allowed the generation of SLF- and GC-deficient bradyzoites that are viable as tachyzoites but show a reduction in cyst burden during the onset of chronic infection. Upon oral infection, SLF-deficient cysts failed to establish infection in mice, suggesting SLF's importance for the natural route of T. gondii infection. IMPORTANCE Toxoplasma gondii is an obligate intracellular parasite of the phylum Apicomplexa. This life-threatening opportunistic pathogen establishes a chronic infection in human and animals that is resistant to immune attacks and chemotherapeutic intervention. The slow-growing parasites persist in tissue cysts that constitute a predominant source of transmission. Host cell invasion and egress are two critical steps of the parasite lytic cycle that are governed by a guanylate cyclase complex conserved across the Apicomplexa. A signaling linked factor is characterized here as an additional component of the complex that not only is essential during acute infection but also plays a pivotal role during natural oral infection with tissue cysts' dissemination and persistence.
Collapse
|
41
|
Yang J, Yang X, Liu A, Li Y, Niu Z, Lyu C, Liang X, Xia N, Cui J, Li M, Wu P, Peng C, Shen B. The beta subunit of AMP-activated protein kinase is critical for cell cycle progression and parasite development in Toxoplasma gondii. Cell Mol Life Sci 2022; 79:532. [PMID: 36205781 PMCID: PMC11802946 DOI: 10.1007/s00018-022-04556-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/30/2022] [Accepted: 09/10/2022] [Indexed: 11/03/2022]
Abstract
Toxoplasma gondii is a widespread eukaryotic pathogen that causes life-threatening diseases in humans and diverse animals. It has a complex life cycle with multiple developmental stages, which are timely adjusted according to growth conditions. But the regulatory mechanisms are largely unknown. Here we show that the AMP-activated protein kinase (AMPK), a key regulator of energy homeostasis in eukaryotes, plays crucial roles in controlling the cell cycle progression and bradyzoite development in Toxoplasma. Deleting the β regulatory subunit of AMPK in the type II strain ME49 caused massive DNA damage and increased spontaneous conversion to bradyzoites (parasites at chronic infection stage), leading to severe growth arrest and reduced virulence of the parasites. Under alkaline stress, all Δampkβ mutants converted to a bradyzoite-like state but the cell division pattern was significantly impaired, resulting in compromised parasite viability. Moreover, we found that phosphorylation of the catalytic subunit AMPKα was greatly increased in alkaline stressed parasites, whereas AMPKβ deletion mutants failed to do so. Phosphoproteomics found that many proteins with predicted roles in cell cycle and cell division regulation were differentially phosphorylated after AMPKβ deletion, under both normal and alkaline stress conditions. Together, these results suggest that the parasite AMPK has critical roles in safeguarding cell cycle progression, and guiding the proper exist of the cell cycle to form mature bradyzoites when the parasites are stressed. Consistent with this model, growth of parasites was not significantly altered when AMPKβ was deleted in a strain that was naturally reluctant to bradyzoite development.
Collapse
Affiliation(s)
- Jichao Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Xuke Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Anqi Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Yaqiong Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Zhipeng Niu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Congcong Lyu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Xiaohan Liang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Ningbo Xia
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Jianmin Cui
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Mingjun Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Ping Wu
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, 201210, People's Republic of China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, 201210, People's Republic of China
| | - Bang Shen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China.
- Key Laboratory of Preventive Medicine in Hubei Province, Wuhan, 430070, Hubei Province, People's Republic of China.
- Hubei Hongshan Laboratory, Wuhan, 430070, Hubei Province, People's Republic of China.
| |
Collapse
|
42
|
de Barros RAM, Torrecilhas AC, Marciano MAM, Mazuz ML, Pereira-Chioccola VL, Fux B. Toxoplasmosis in Human and Animals Around the World. Diagnosis and Perspectives in the One Health Approach. Acta Trop 2022; 231:106432. [PMID: 35390311 DOI: 10.1016/j.actatropica.2022.106432] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/25/2022] [Accepted: 03/27/2022] [Indexed: 12/12/2022]
Abstract
Toxoplasmosis is a unique health disease that significantly affects the health of humans, domestic animals, wildlife and is present in ecosystems, including water, soil and food. Toxoplasma gondii is one of the best-adapted parasites in the word. This parasite is able to persist for long periods in its hosts, in different geographic regions of the word. This review summarizes the current literature of these themes, focusing on: (1) toxoplasmosis, a zoonotic infection; (2) One health approach and toxoplasmosis; (3) human toxoplasmosis; (4) animal toxoplasmosis; (5) toxoplasmosis diagnosis, as immunological, parasitological and molecular diagnosis; (6) T. gondii outbreaks caused by infected meat, milk and dairy products, as well as, vegetables and water consume; (7) studies in experimental models; (8) genetic characterization of T. gondii strains; (9) extracellular vesicles and miRNA; and (10) future perspectives on T. gondii and toxoplasmosis. The vast prevalence of toxoplasmosis in both humans and animals and the dispersion and resistence of T. gondii parasites in environment highlight the importance of the one health approach in diagnostic and control of the disease. Here the different aspects of the one health approach are presented and discussed.
Collapse
Affiliation(s)
- Rosangela Aparecida Müller de Barros
- Unidade de Medicina Tropical, Departamento de Patologia, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil.; Programa em Doenças Infecciosas, Centro de Doenças Infecciosas, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil..
| | - Ana Claudia Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), Campus Diadema, Sao Paulo, SP, Brazil..
| | | | - Monica Leszkowicz Mazuz
- Parasitology Division, Kimron Veterinary Institute, Israeli Veterinary Service and Animal Health, Ministry of Agriculture and Rural Development Beit Dagan, 5025000, Israel..
| | | | - Blima Fux
- Unidade de Medicina Tropical, Departamento de Patologia, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil.; Programa em Doenças Infecciosas, Centro de Doenças Infecciosas, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil..
| |
Collapse
|
43
|
Wu M, An R, Zhou N, Chen Y, Cai H, Yan Q, Wang R, Luo Q, Yu L, Chen L, Du J. Toxoplasma gondii CDPK3 Controls the Intracellular Proliferation of Parasites in Macrophages. Front Immunol 2022; 13:905142. [PMID: 35757711 PMCID: PMC9226670 DOI: 10.3389/fimmu.2022.905142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/16/2022] [Indexed: 11/30/2022] Open
Abstract
Interferon-γ (IFN-γ)-activated macrophages restrain the replication of intracellular parasites and disrupt the integrity of vacuolar pathogens. The growth of the less virulent type II strain of Toxoplasma gondii (such as ME49) was strongly inhibited by IFN-γ-activated murine macrophages. However, the mechanism of resistance is poorly understood. Immunity-related GTPases (IRGs) as well as guanylate-binding proteins (GBPs) contributed to this antiparasitic effect. Previous studies showed the cassette of autophagy-related proteins including Atg7, Atg3, and Atg12-Atg5-Atg16L1 complex, plays crucial roles in the proper targeting of IFN-γ effectors onto the parasitophorous vacuole (PV) membrane of Toxoplasma gondii and subsequent control of parasites. TgCDPK3 is a calcium dependent protein kinase, located on the parasite periphery, plays a crucial role in parasite egress. Herein, we show that the less virulent strain CDPK3 (ME49, type II) can enhance autophagy activation and interacts with host autophagy proteins Atg3 and Atg5. Infection with CDPK3-deficient ME49 strain resulted in decreased localization of IRGs and GBPs around PV membrane. In vitro proliferation and plaque assays showed that CDPK3-deficient ME49 strain replicated significantly more quickly than wild-type parasites. These data suggested that TgCDPK3 interacts with the host Atg3 and Atg5 to promote the localization of IRGs and GBPs around PV membrane and inhibits the intracellular proliferation of parasites, which is beneficial to the less virulent strain of Toxoplasma gondii long-term latency in host cells.
Collapse
Affiliation(s)
- Minmin Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Provincial Key Laboratory of Zoonoses of High Institutions of Anhui, Anhui Medical University, Hefei, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, China
| | - Ran An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Provincial Key Laboratory of Zoonoses of High Institutions of Anhui, Anhui Medical University, Hefei, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, China
| | - Nan Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Provincial Key Laboratory of Zoonoses of High Institutions of Anhui, Anhui Medical University, Hefei, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, China
| | - Ying Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Provincial Key Laboratory of Zoonoses of High Institutions of Anhui, Anhui Medical University, Hefei, China.,School of Nursing, Anhui Medical University, Hefei, China
| | - Haijian Cai
- The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Provincial Key Laboratory of Zoonoses of High Institutions of Anhui, Anhui Medical University, Hefei, China
| | - Qi Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Provincial Key Laboratory of Zoonoses of High Institutions of Anhui, Anhui Medical University, Hefei, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, China
| | - Ru Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Provincial Key Laboratory of Zoonoses of High Institutions of Anhui, Anhui Medical University, Hefei, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, China
| | - Qingli Luo
- The Provincial Key Laboratory of Zoonoses of High Institutions of Anhui, Anhui Medical University, Hefei, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, China
| | - Li Yu
- The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Provincial Key Laboratory of Zoonoses of High Institutions of Anhui, Anhui Medical University, Hefei, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, China
| | - Lijian Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jian Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Research Center for Infectious Diseases, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,The Provincial Key Laboratory of Zoonoses of High Institutions of Anhui, Anhui Medical University, Hefei, China.,The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, Hefei, China
| |
Collapse
|
44
|
Martín-Escolano J, Marín C, Rosales MJ, Tsaousis AD, Medina-Carmona E, Martín-Escolano R. An Updated View of the Trypanosoma cruzi Life Cycle: Intervention Points for an Effective Treatment. ACS Infect Dis 2022; 8:1107-1115. [PMID: 35652513 PMCID: PMC9194904 DOI: 10.1021/acsinfecdis.2c00123] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Chagas disease (CD)
is a parasitic, systemic, chronic, and often
fatal illness caused by infection with the protozoan Trypanosoma
cruzi. The World Health Organization classifies CD as the
most prevalent of poverty-promoting neglected tropical diseases, the
most important parasitic one, and the third most infectious disease
in Latin America. Currently, CD is a global public health issue that
affects 6–8 million people. However, the current approved treatments
are limited to two nitroheterocyclic drugs developed more than 50
years ago. Many efforts have been made in recent decades to find new
therapies, but our limited understanding of the infection process,
pathology development, and long-term nature of this disease has made
it impossible to develop new drugs, effective treatment, or vaccines.
This Review aims to provide a comprehensive update on our understanding
of the current life cycle, new morphological forms, and genetic diversity
of T. cruzi, as well as identify intervention points
in the life cycle where new drugs and treatments could achieve a parasitic
cure.
Collapse
Affiliation(s)
- Javier Martín-Escolano
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E41013 Seville, Spain
| | - Clotilde Marín
- Department of Parasitology, University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| | - María J. Rosales
- Department of Parasitology, University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| | - Anastasios D. Tsaousis
- Laboratory of Molecular & Evolutionary Parasitology, RAPID group, School of Biosciences, University of Kent, Canterbury CT2 7NJ, U.K
| | - Encarnación Medina-Carmona
- Department of Physical Chemistry, University of Granada, 18071 Granada, Spain
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, U.K
| | - Rubén Martín-Escolano
- Laboratory of Molecular & Evolutionary Parasitology, RAPID group, School of Biosciences, University of Kent, Canterbury CT2 7NJ, U.K
| |
Collapse
|
45
|
Barakat AM, El Fadaly HAM, Selem RF, Madboli AENA, Abd El-Razik KA, Hassan EA, Alghamdi AH, Elmahallawy EK. Tamoxifen Increased Parasite Burden and Induced a Series of Histopathological and Immunohistochemical Changes During Chronic Toxoplasmosis in Experimentally Infected Mice. Front Microbiol 2022; 13:902855. [PMID: 35707167 PMCID: PMC9189418 DOI: 10.3389/fmicb.2022.902855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
The global distribution of breast cancer and the opportunistic nature of the parasite have resulted in many patients with breast cancer becoming infected with toxoplasmosis. However, very limited information is available about the potential effects of tamoxifen on chronic toxoplasmosis and its contribution to the reactivation of the latent infection. The present study investigated the potential effects of tamoxifen on chronic toxoplasmosis in animal models (Swiss albino mice). Following induction of chronic toxoplasmosis and treatment with the drug for 14 and 28 days, the anti-parasitic effects of tamoxifen were evaluated by parasitological assessment and counting of Toxoplasma cysts. In addition, the effects of the drug on the parasite load were evaluated and quantitated using TaqMan real-time quantitative PCR followed by investigation of the major histopathological changes and immunohistochemical findings. Interestingly, tamoxifen increased the parasite burden on animals treated with the drug during 14 and 28 days as compared with the control group. The quantification of the DNA concentrations of Toxoplasma P29 gene after the treatment with the drug revealed a higher parasite load in both treated groups vs. control groups. Furthermore, treatment with tamoxifen induced a series of histopathological and immunohistochemical changes in the kidney, liver, brain, and uterus, revealing the exacerbating effect of tamoxifen against chronic toxoplasmosis. These changes were represented by the presence of multiple T. gondii tissue cysts in the lumen of proximal convoluted tubules associated with complete necrosis in their lining epithelium of the kidney section. Meanwhile, liver tissue revealed multiple T. gondii tissue cysts in hepatic parenchyma which altered the structure of hepatocytes. Moreover, clusters of intracellular tachyzoites were observed in the lining epithelium of endometrium associated with severe endometrial necrosis and appeared as diffuse nuclear pyknosis combined with sever mononuclear cellular infiltration. Brain tissues experienced the presence of hemorrhages in pia mater and multiple T. gondii tissue cysts in brain tissue. The severity of the lesions was maximized by increasing the duration of treatment. Collectively, the study concluded novel findings in relation to the potential role of tamoxifen during chronic toxoplasmosis. These findings are very important for combating the disease, particularly in immunocompromised patients which could be life-threatening.
Collapse
Affiliation(s)
- Ashraf Mohamed Barakat
- Department of Zoonotic Diseases, Veterinary Research Institute, National Research Centre, Giza, Egypt
- *Correspondence: Ashraf Mohamed Barakat,
| | | | - Rabab Fawzy Selem
- Department of Parasitology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Abd El-Nasser A. Madboli
- Department of Animal Reproduction, Veterinary Research Institute, National Research Centre, Giza, Egypt
| | - Khaled A. Abd El-Razik
- Department of Animal Reproduction, Veterinary Research Institute, National Research Centre, Giza, Egypt
| | - Ehssan Ahmed Hassan
- Department of Biology, College of Science and Humanities, Prince Sattam bin Abdul Aziz University, Alkharj, Saudi Arabia
- Department of Zoology, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Ali H. Alghamdi
- Department of Biology, Faculty of Science, Albaha University, Alaqiq, Saudi Arabia
| | - Ehab Kotb Elmahallawy
- Department of Zoonoses, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
- Ehab Kotb Elmahallawy,
| |
Collapse
|
46
|
Host cell proteins modulated upon Toxoplasma infection identified using proteomic approaches: a molecular rationale. Parasitol Res 2022; 121:1853-1865. [PMID: 35552534 DOI: 10.1007/s00436-022-07541-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/12/2022] [Indexed: 10/18/2022]
Abstract
Toxoplasma gondii is a pathogenic protozoan parasite belonging to the apicomplexan phylum that infects the nucleated cells of warm-blooded hosts leading to an infectious disease known as toxoplasmosis. Apicomplexan parasites such as T. gondii can display different mechanisms to control or manipulate host cells signaling at different levels altering the host subcellular genome and proteome. Indeed, Toxoplasma is able to modulate host cell responses (especially immune responses) during infection to its advantage through both structural and functional changes in the proteome of different infected cells. Consequently, parasites can transform the invaded cells into a suitable environment for its own replication and the induction of infection. Proteomics as an applicable tool can identify such critical proteins involved in pathogen (Toxoplasma)-host cell interactions and consequently clarify the cellular mechanisms that facilitate the entry of pathogens into host cells, and their replication and transmission, as well as the central mechanisms of host defense against pathogens. Accordingly, the current paper reviews several proteins (identified using proteomic approaches) differentially expressed in the proteome of Toxoplasma-infected host cells (macrophages and human foreskin fibroblasts) and tissues (brain and liver) and highlights their plausible functions in the cellular biology of the infected cells. The identification of such modulated proteins and their related cell impact (cell responses/signaling) can provide further information regarding parasite pathogenesis and biology that might lead to a better understanding of therapeutic strategies and novel drug targets.
Collapse
|
47
|
Gubbels MJ, Ferguson DJP, Saha S, Romano JD, Chavan S, Primo VA, Michaud C, Coppens I, Engelberg K. Toxoplasma gondii's Basal Complex: The Other Apicomplexan Business End Is Multifunctional. Front Cell Infect Microbiol 2022; 12:882166. [PMID: 35573773 PMCID: PMC9103881 DOI: 10.3389/fcimb.2022.882166] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/30/2022] [Indexed: 01/08/2023] Open
Abstract
The Apicomplexa are famously named for their apical complex, a constellation of organelles at their apical end dedicated to invasion of their host cells. In contrast, at the other end of the cell, the basal complex (BC) has been overshadowed since it is much less prominent and specific functions were not immediately obvious. However, in the past decade a staggering array of functions have been associated with the BC and strides have been made in understanding its structure. Here, these collective insights are supplemented with new data to provide an overview of the understanding of the BC in Toxoplasma gondii. The emerging picture is that the BC is a dynamic and multifunctional complex, with a series of (putative) functions. The BC has multiple roles in cell division: it is the site where building blocks are added to the cytoskeleton scaffold; it exerts a two-step stretch and constriction mechanism as contractile ring; and it is key in organelle division. Furthermore, the BC has numerous putative roles in 'import', such as the recycling of mother cell remnants, the acquisition of host-derived vesicles, possibly the uptake of lipids derived from the extracellular medium, and the endocytosis of micronemal proteins. The latter process ties the BC to motility, whereas an additional role in motility is conferred by Myosin C. Furthermore, the BC acts on the assembly and/or function of the intravacuolar network, which may directly or indirectly contribute to the establishment of chronic tissue cysts. Here we provide experimental support for molecules acting in several of these processes and identify several new BC proteins critical to maintaining the cytoplasmic bridge between divided parasites. However, the dispensable nature of many BC components leaves many questions unanswered regarding its function. In conclusion, the BC in T. gondii is a dynamic and multifunctional structure at the posterior end of the parasite.
Collapse
Affiliation(s)
- Marc-Jan Gubbels
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - David J. P. Ferguson
- Nuffield Department of Clinical Laboratory Science, University of Oxford John Radcliffe Hospital, Oxford, United Kingdom
- Department of Biological and Medical Sciences, Faculty of Health and Life Science, Oxford Brookes University, Oxford, United Kingdom
| | - Sudeshna Saha
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Julia D. Romano
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Suyog Chavan
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Vincent A. Primo
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Cynthia Michaud
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Klemens Engelberg
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| |
Collapse
|
48
|
Sasai M, Yamamoto M. Anti-toxoplasma host defense systems and the parasitic counterdefense mechanisms. Parasitol Int 2022; 89:102593. [DOI: 10.1016/j.parint.2022.102593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 04/12/2022] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
|
49
|
Toxoplasma gondii infection/exposure and the risk of brain tumors: A systematic review and meta-analysis. Cancer Epidemiol 2022; 77:102119. [DOI: 10.1016/j.canep.2022.102119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/27/2022] [Accepted: 02/03/2022] [Indexed: 01/10/2023]
|
50
|
Soares GLDS, Leão ERLPD, Freitas SF, Alves RMC, Tavares NDP, Costa MVN, Menezes GCD, Oliveira JHPD, Guerreiro LCF, Assis ACLD, Araújo SC, Franco FTDC, Anaissi AKM, Carmo ELD, Morais RDAPB, Demachki S, Diniz JAP, Nunes HM, Anthony DC, Diniz DG, Diniz CWP. Behavioral and Neuropathological Changes After Toxoplasma gondii Ocular Conjunctival Infection in BALB/c Mice. Front Cell Infect Microbiol 2022; 12:812152. [PMID: 35372100 PMCID: PMC8965508 DOI: 10.3389/fcimb.2022.812152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/07/2022] [Indexed: 11/15/2022] Open
Abstract
Ocular infection with Toxoplasma gondii causes toxoplasmosis in mice. However, following ocular infection with tachyzoites, the cause of the accompanying progressive changes in hippocampal-dependent tasks, and their relationship with the morphology and number of microglia, is less well understood. Here, in 6-month-old, female BALB/c mice, 5 μl of a suspension containing 48.5 × 106 tachyzoites/ml was introduced into the conjunctival sac; control received an equal volume of saline. Before and after instillation, all mice were subject to an olfactory discrimination (OD) test, using predator (cat) feces, and to an open-field (OF) task. After the behavioral tests, the animals were culled at either 22 or 44 days post-instillation (dpi), and the brains and retinas were dissected and processed for immunohistochemistry. The total number of Iba-1-immunolabeled microglia in the molecular layer of the dentate gyrus was estimated, and three-dimensional reconstructions of the cells were evaluated. Immobility was increased in the infected group at 12, 22, and 43 dpi, but the greatest immobility was observed at 22 dpi and was associated with reduced line crossing in the OF and distance traveled. In the OD test, infected animals spent more time in the compartment with feline fecal material at 14 and at 43 dpi. No OD changes were observed in the control group. The number of microglia was increased at 22 dpi but returned to control levels by 44 dpi. These changes were associated with the differentiation of T. gondii tachyzoites into bradyzoite-enclosed cysts within the brain and retina. Thus, infection of mice with T. gondii alters exploratory behavior, gives rise to a loss in predator’s odor avoidance from 2 weeks after infection, increased microglia number, and altered their morphology in the molecular layer of the dentate gyrus.
Collapse
|