1
|
Lem M, Rh H, Dg B, Barkhouse A, Miller DW, Raun N, Sa A. The caterpillar Manduca sexta brain shows changes in gene expression and protein abundance correlating with parasitic manipulation of behaviour. Sci Rep 2024; 14:31773. [PMID: 39738473 DOI: 10.1038/s41598-024-82506-4] [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: 07/03/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
The parasitic wasp, Cotesia congregata, manipulates the behaviour of its host, the caterpillar Manduca sexta. The female wasp injects her eggs and a symbiotic virus (i.e. bracovirus, CcBV) into the body of its host. The host's behaviour remains unchanged until the wasps exit the caterpillar, and then the caterpillar becomes a non-feeding "bodyguard" for the wasp cocoons. Using proteomic, transcriptomic and qPCR studies, we discovered an increase in antimicrobial peptide gene expression and protein abundance in the host central nervous system at the time of wasp emergence, correlating with the change in host behaviour. These results support the hypothesis that the wasps hyperactivate an immune-neural connection to help create the change in behaviour. At the time of wasp emergence, there was also an increase in bracoviral gene expression and proteins in the host brain, suggesting that the bracovirus may also be involved in altering host behaviour. Other changes in gene expression and protein abundance suggest that synaptic transmission may be altered after wasp emergence, and a reduction in descending neural activity from the host's brain provides indirect support for this hypothesis.
Collapse
Affiliation(s)
- McMillan Lem
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Herbison Rh
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Biron Dg
- Lab Microorganismes: Génome et Environment, Université Clermont Auvergne, UMR CNRS, Paris, 6023, France
| | - A Barkhouse
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - D W Miller
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - N Raun
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, B3H 4R2, Canada
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, 6525 GA, the Netherlands
| | - Adamo Sa
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada.
| |
Collapse
|
2
|
Sowunmi AA, Omeiza NA, Bakre A, Abdulrahim HA, Aderibigbe AO. Dissecting the antidepressant effect of troxerutin: modulation of neuroinflammatory and oxidative stress biomarkers in lipopolysaccharide-treated mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9965-9979. [PMID: 38951153 DOI: 10.1007/s00210-024-03252-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024]
Abstract
The role of neuroinflammation in the pathogenesis of depression has prompted the search for new antidepressants. Troxerutin, a bioflavonoid with anti-inflammatory and antioxidant properties, has shown promise, but its impact on neurobehavioral functions remains poorly understood. This study aimed to investigate the antidepressant potential of troxerutin and its effect on the neuroinflammatory response. Here, we exposed male Swiss mice (n = 5/group) to various treatments, including naive and negative controls receiving distilled water, troxerutin-treated groups administered at different doses (10, 20, 40 mg/kg, i.p.), and an imipramine-treated group (25 mg/kg, i.p.). After seven days of treatment, with the exception of the naive group, mice were administered a single dose of lipopolysaccharide (LPS, 0.83 mg/kg). Behavioral evaluations, consisting of the novelty-suppressed feeding (NSF) test, forced swim test (FST), and open field test (OFT), were conducted. Additionally, brain samples were collected for biochemical and immunohistochemical analyses. Troxerutin significantly reduced immobility time in the FST and mitigated behavioral deficits in the NSF test. Additionally, troxerutin increased glutathione (GSH) and superoxide dismutase (SOD) levels while reducing nitrite, malondialdehyde (MDA), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interferon-gamma (IFN-γ) levels compared to the negative control. Immunohistochemistry analysis revealed decreased expression of inducible nitric oxide synthase (iNOS) and nuclear factor-kappa B (NF-κB) in troxerutin-treated mice. Overall, these findings suggest that troxerutin exerts significant antidepressive-like effects, likely mediated by its anti-inflammatory and antioxidant mechanisms. The reduction in neuroinflammatory and oxidative stress biomarkers, along with the improvement in behavioral outcomes, underscores troxerutin's potential as a therapeutic agent for depression.
Collapse
Affiliation(s)
- Abimbola A Sowunmi
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Noah A Omeiza
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria.
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, Academia Sinica, Taipei, Taiwan.
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Adewale Bakre
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Halimat A Abdulrahim
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Adegbuyi O Aderibigbe
- Department of Pharmacology and Therapeutics, Neuropharmacology Unit, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| |
Collapse
|
3
|
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
|
4
|
Baker TL, Wright DK, Uboldi AD, Tonkin CJ, Vo A, Wilson T, McDonald SJ, Mychasiuk R, Semple BD, Sun M, Shultz SR. A pre-existing Toxoplasma gondii infection exacerbates the pathophysiological response and extent of brain damage after traumatic brain injury in mice. J Neuroinflammation 2024; 21:14. [PMID: 38195485 PMCID: PMC10775436 DOI: 10.1186/s12974-024-03014-w] [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: 10/21/2023] [Accepted: 01/04/2024] [Indexed: 01/11/2024] Open
Abstract
Traumatic brain injury (TBI) is a key contributor to global morbidity that lacks effective treatments. Microbial infections are common in TBI patients, and their presence could modify the physiological response to TBI. It is estimated that one-third of the human population is incurably infected with the feline-borne parasite, Toxoplasma gondii, which can invade the central nervous system and result in chronic low-grade neuroinflammation, oxidative stress, and excitotoxicity-all of which are also important pathophysiological processes in TBI. Considering the large number of TBI patients that have a pre-existing T. gondii infection prior to injury, and the potential mechanistic synergies between the conditions, this study investigated how a pre-existing T. gondii infection modified TBI outcomes across acute, sub-acute and chronic recovery in male and female mice. Gene expression analysis of brain tissue found that neuroinflammation and immune cell markers were amplified in the combined T. gondii + TBI setting in both males and females as early as 2-h post-injury. Glutamatergic, neurotoxic, and oxidative stress markers were altered in a sex-specific manner in T. gondii + TBI mice. Structural MRI found that male, but not female, T. gondii + TBI mice had a significantly larger lesion size compared to their uninfected counterparts at 18-weeks post-injury. Similarly, diffusion MRI revealed that T. gondii + TBI mice had exacerbated white matter tract abnormalities, particularly in male mice. These novel findings indicate that a pre-existing T. gondii infection affects the pathophysiological aftermath of TBI in a sex-dependent manner, and may be an important modifier to consider in the care and prognostication of TBI patients.
Collapse
Affiliation(s)
- Tamara L Baker
- Department of Neuroscience, Central Clinical School, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Alessandro D Uboldi
- Division of Infectious Disease and Immune Defense, , The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Christopher J Tonkin
- Division of Infectious Disease and Immune Defense, , The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Anh Vo
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Trevor Wilson
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.
- Health Sciences, Vancouver Island University, Nanaimo, BC, Canada.
| |
Collapse
|
5
|
Wakid MH, Alsulami MN, Farid M, El Kholy WA. Potential Anti-Toxoplasmosis Efficiency of Phoenix dactylifera Extracts Loaded on Selenium Nanoparticles. Infect Drug Resist 2023; 16:7743-7758. [PMID: 38144223 PMCID: PMC10749168 DOI: 10.2147/idr.s443047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023] Open
Abstract
Background Toxoplasmosis is a parasitic disease caused by Toxoplasma gondii that infects humans and many types of mammals and birds. Objective To investigate the effect of selenium nanoparticles (SeNPs) and Phoenix dactylifera (Pd) extracts loaded on SeNPs as a new agent to combat chronic T. gondii infections in murine model as an alternative method to standard Spiramycin drug therapy. Methods A total of 64 female mice were randomly divided into eight groups: GI: Normal control, GII: Positive control, GIII: infected and treated with Spiramycin, GIV: infected and treated with SeNPs, GV: infected and treated with aqueous extract of Pd, GVI: infected and treated with methanolic extract of Pd, GVII: infected and treated with aqueous extract of Pd loaded on SeNPs, GVIII: infected and treated with methanolic extract of Pd loaded on SeNPs. Date palm (P. dactylifera) fruits were identified and collected from the farms of Saudi Arabia. Preparation and characterization of SeNPs were done. The parasitological, histopathological examinations and biochemical changes were evaluated in all groups. Results Parasitological results showed significant differences in GVII in comparison to GII while GVIII showed significant differences in comparison to GII and GIII. The histopathological section of the cerebral cortex showed obvious alterations in the infected compared with untreated control groups. Aqueous and methanolic extracts of P. dactylifera loaded on SeNPs treatment showed improvement that indicated by few perivascular cuffing with few inflammatory cell infiltrations. Few granule cells with mild intracellular vacuolation and edema few deformed neurons with deep pyknotic nuclei. Microglia cells expression of Iba-1 and inflammatory cytokines (IL-4, IL-10 and INF-γ) in serum of all groups was higher in GII and lowest in GVIII followed by GVII. Conclusion SeNPs and P. dactylifera extracts loaded on SeNPs could be a potent agent to combat T. gondii infections.
Collapse
Affiliation(s)
- Majed H Wakid
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muslimah N Alsulami
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Mohamed Farid
- Sciences Academy of Experimental Researches, Special Scientific Foundation, Mansoura, Egypt
| | - Walaa A El Kholy
- Department of Parasitology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
6
|
Liu S, Yan Z, Peng Y, Liu Y, Li Y, Xu D, Gong Y, Cui Z, Wu Y, Zhang Y, Wang D, Pan W, Yang X. Lentinan has a beneficial effect on cognitive deficits induced by chronic Toxoplasma gondii infection in mice. Parasit Vectors 2023; 16:454. [PMID: 38093309 PMCID: PMC10717010 DOI: 10.1186/s13071-023-06023-5] [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: 04/29/2023] [Accepted: 10/19/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Toxoplasma gondii (T. gondii) is increasingly considered a risk factor for neurodegenerative diseases. However, there is only limited information on the development of drugs for T. gondii infection. Lentinan from Lentinula edodes is a bioactive ingredient with the potential to enhance anti-infective immunity. The present study aimed to investigate the neuroprotective effect of lentinan on T. gondii-associated cognitive deficits in mice. METHODS A chronic T. gondii infection mouse model was established by administering 10 cysts of T. gondii by gavage. Lentinan was intraperitoneally administered 2 weeks before infection. Behavioral tests, RNA sequencing, immunofluorescence, transmission electron microscopy and Golgi-Cox staining were performed to assess the effect of lentinan on cognitive deficits and neuropathology in vivo. In vitro, the direct and indirect effects of lentinan on the proliferation of T. gondii tachyzoites were evaluated in the absence and presence of BV-2 cells, respectively. RESULTS Lentinan prevented T. gondii-induced cognitive deficits and altered the transcriptome profile of genes related to neuroinflammation, microglial activation, synaptic function, neural development and cognitive behavior in the hippocampus of infected mice. Moreover, lentinan reduced the infection-induced accumulation of microglia and downregulated the mRNA expression of proinflammatory cytokines. In addition, the neurite and synaptic ultrastructural damage in the hippocampal CA1 region due to infection was ameliorated by lentinan administration. Lentinan decreased the cyst burden in the brains of infected mice, which was correlated with behavioral performance. In line with this finding, lentinan could significantly inhibit the proliferation of T. gondii tachyzoites in the microglial cell line BV2, although lentinan had no direct inhibitory effect on parasite growth. CONCLUSIONS Lentinan prevents cognitive deficits via the improvement of neurite impairment and synaptic loss induced by T. gondii infection, which may be associated with decreased cyst burden in the brain. Overall, our findings indicate that lentinan can ameliorate T. gondii-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuxi Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ziyi Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuan Peng
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yunqiu Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yiling Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Daxiang Xu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yuying Gong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Zeyu Cui
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yongshui Wu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yumei Zhang
- Department of Pathogenic Biology, Binzhou Medical University, Binzhou, 256603, Shandong, China
| | - Dahui Wang
- Liangshan College (Li Shui) China, Lishui University, Lishui, 323000, Zhejiang, China.
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
7
|
Ding K, Xu Q, Zhang X, Liu S. Metabolomic insights into neurological effects of BDE-47 exposure in the sea cucumber Apostichopus japonicus. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 266:115558. [PMID: 37820477 DOI: 10.1016/j.ecoenv.2023.115558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/21/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
The persistent organic pollutant 2,2',4,4'-Tetrabromodiphenyl ether (BDE-47), a prevalent congener among polybrominated diphenyl ethers (PBDEs), exhibits potent bioaccumulation and toxicity. Despite extensive research into the adverse effects of BDE-47, its neurotoxicity in sea cucumbers remains unexplored. Given the crucial role of the sea cucumber's nervous system in survival and adaptation, evaluating the impacts of BDE-47 is vital for sustainable aquaculture and consumption. In this study, we employed ultra-high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Triple-TOF-MS) to analyze metabolomic changes in neuro-related tissues of Apostichopus japonicus exposed to low (0.1 µg/L), medium (1.0 µg/L), and high (10.0 µg/L) BDE-47 concentrations. We identified significantly changed metabolites in each exposure group (87 in low, 79 in medium, and 102 in high), affecting a variety of physiological processes such as steroid hormone balance, nucleotide metabolism, energy metabolism, neurotransmitter levels, and neuroprotection. In addition, we identified concentration-dependent, common, and some other metabolic responses in the neuro-related tissues. Our findings reveal critical insights into the neurotoxic effects of BDE-47 in sea cucumbers and contribute to risk assessment related to BDE-47 exposure in the sea cucumber industry, paving the way for future neurotoxicological research in invertebrates.
Collapse
Affiliation(s)
- Kui Ding
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao 266061, China; Laboratory of Marine Ecology and Environmental Science, National Laboratory for Marine Science and Technology, Qingdao 266061, China; CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China.
| | - Qinzeng Xu
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao 266061, China; Laboratory of Marine Ecology and Environmental Science, National Laboratory for Marine Science and Technology, Qingdao 266061, China
| | - Xuelei Zhang
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao 266061, China; Laboratory of Marine Ecology and Environmental Science, National Laboratory for Marine Science and Technology, Qingdao 266061, China
| | - Shilin Liu
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| |
Collapse
|
8
|
Anaya-Martínez V, Anacleto-Santos J, Mondragón-Flores R, Zepeda-Rodríguez A, Casarrubias-Tabarez B, de Jesús López-Pérez T, de Alba-Alvarado MC, Martínez-Ortiz-de-Montellano C, Carrasco-Ramírez E, Rivera-Fernández N. Changes in the Proliferation of the Neural Progenitor Cells of Adult Mice Chronically Infected with Toxoplasma gondii. Microorganisms 2023; 11:2671. [PMID: 38004683 PMCID: PMC10673519 DOI: 10.3390/microorganisms11112671] [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: 10/05/2023] [Revised: 10/25/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023] Open
Abstract
During Toxoplasma gondii chronic infection, certain internal factors that trigger the proliferation of neural progenitor cells (NPCs), such as brain inflammation, cell death, and changes in cytokine levels, are observed. NPCs give rise to neuronal cell types in the adult brain of some mammals. NPCs are capable of dividing and differentiating into a restricted repertoire of neuronal and glial cell types. In this study, the proliferation of NPCs was evaluated in CD-1 adult male mice chronically infected with the T. gondii ME49 strain. Histological brain sections from the infected mice were evaluated in order to observe T. gondii tissue cysts. Sagittal and coronal sections from the subventricular zone of the lateral ventricles and from the subgranular zone of the hippocampal dentate gyrus, as well as sagittal sections from the rostral migratory stream, were obtained from infected and non-infected mice previously injected with bromodeoxyuridine (BrdU). A flotation immunofluorescence technique was used to identify BrdU+ NPC. The scanning of BrdU+ cells was conducted using a confocal microscope, and the counting was performed with ImageJ® software (version 1.48q). In all the evaluated zones from the infected mice, a significant proliferation of the NPCs was observed when compared with that of the control group. We concluded that chronic infection with T. gondii increased the proliferation of NPCs in the three evaluated zones. Regardless of the role these cells are playing, our results could be useful to better understand the pathogenesis of chronic toxoplasmosis.
Collapse
Affiliation(s)
- Verónica Anaya-Martínez
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac, Lomas Anáhuac, Naucalpan de Juárez 52786, Estado de México, Mexico;
| | - Jhony Anacleto-Santos
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Ciudad de México 04510, Mexico; (J.A.-S.); (T.d.J.L.-P.); (M.C.d.A.-A.); (E.C.-R.)
| | | | - Armando Zepeda-Rodríguez
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Ciudad de México 04510, Mexico; (A.Z.-R.); (B.C.-T.)
| | - Brenda Casarrubias-Tabarez
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Ciudad de México 04510, Mexico; (A.Z.-R.); (B.C.-T.)
| | - Teresa de Jesús López-Pérez
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Ciudad de México 04510, Mexico; (J.A.-S.); (T.d.J.L.-P.); (M.C.d.A.-A.); (E.C.-R.)
| | - Mariana Citlalli de Alba-Alvarado
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Ciudad de México 04510, Mexico; (J.A.-S.); (T.d.J.L.-P.); (M.C.d.A.-A.); (E.C.-R.)
| | - Cintli Martínez-Ortiz-de-Montellano
- Departamento de Parasitología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico;
| | - Elba Carrasco-Ramírez
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Ciudad de México 04510, Mexico; (J.A.-S.); (T.d.J.L.-P.); (M.C.d.A.-A.); (E.C.-R.)
| | - Norma Rivera-Fernández
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Ciudad de México 04510, Mexico; (J.A.-S.); (T.d.J.L.-P.); (M.C.d.A.-A.); (E.C.-R.)
| |
Collapse
|
9
|
Social interaction, psychotic disorders and inflammation: A triangle of interest. Prog Neuropsychopharmacol Biol Psychiatry 2023; 122:110697. [PMID: 36521587 DOI: 10.1016/j.pnpbp.2022.110697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Social interaction difficulties are a hallmark of psychotic disorders, which in some cases can be definitely traced back to autoimmunological causes. Interestingly, systemic and intrathecal inflammation have been shown to significantly influence social processing by increasing sensitivity to threatening social stimuli, which bears some resemblance to psychosis. In this article, we review evidence for the involvement of systemic and intrathecal inflammatory processes in psychotic disorders and how this might help to explain some of the social impairments associated with this group of disorders. Vice versa, we also discuss evidence for the immunomodulatory function of social interactions and their potential role for therapeutic interventions in psychotic disorders.
Collapse
|
10
|
Wana MN, Watanabe M, Chiroma SM, Unyah NZ, Abdullahi SA, Nordin S, Basir R, Mohd Moklas MA, Majid RA. Toxoplasma gondii induced cognitive impairment in rats via dysregulation of dopamine receptors and indoleamine 2,3 dioxygenase. Heliyon 2023; 9:e14370. [PMID: 36950587 PMCID: PMC10025920 DOI: 10.1016/j.heliyon.2023.e14370] [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: 03/05/2022] [Revised: 02/17/2023] [Accepted: 03/01/2023] [Indexed: 03/18/2023] Open
Abstract
Toxoplasma gondii (T. gondii) is a parasite capable of residing in the brain of their host which influences behaviour changes due to alterations in the neurotransmitters. Consequently, dopamine receptors (DRD) and indoleamine 2, 3 dioxygenase (IDO) dysregulation facilitate the progression of behaviour changes in a host as a response to infection. This study tested the effect of neurotransmitter changes as a result of T. gondii infection on rats cognitive impairment. The T. gondii strain of type I, II and III from Malaysia were previously identified by standard procedures. Sporulated oocysts each of type I, II and III were inoculated separately into three groups of Wistar rats (n = 9) respectively. Two separate control groups received either phosphate buffered saline (PBS) or MK-801 (dizocilpine). Behaviour changes were evaluated at nine weeks post infection in a square box, elevated plus maze and gene expression level of DRD and IDO compounds. The study revealed increased fatal feline attraction, reduced anxiety, decreased DRD and increased IDO gene expression in the T. gondii infected groups and MK-801 compared to the PBS control group. In conclusion, T. gondii infection alter the level of neurotransmitters in rat which cause cognitive impairment. This implies that all the T. gondii strain can cause behaviour changes if human were infected.
Collapse
Affiliation(s)
- Mohammed Nasiru Wana
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Department of Biological Sciences, Faculty of Science, Abubakar Tafawa Balewa University Bauchi, Nigeria
| | - Malaika Watanabe
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Samaila Musa Chiroma
- Department of Human Anatomy, Faculty of Basic Medical Sciences, University of Maiduguri, Nigeria
- Newcastle University Medicine Malaysia (NuMed) No 1, Jalan Sarjana 1,Kota Ilmu, EduCity@Iskandar,79200 Iskandar Puteri (formerly Nusajaya) Johor-Malaysia
| | - Ngah Zasmy Unyah
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Sharif Alhassan Abdullahi
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Department of Medical Microbiology and Parasitology, Faculty of Clinical Sciences, Bayero University, Kano, Nigeria
| | - Shariza Nordin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Rusliza Basir
- Department of Human Anatomy, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Mohamad Aris Mohd Moklas
- Department of Human Anatomy, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Corresponding author.
| | - Roslaini Abd Majid
- Department of Pre-Clinical, Faculty of Medicine and Defence Health, National Defence University of Malaysia, Kem Sungai Besi, 57000, Kuala Lumpur, Malaysia
- Corresponding author.
| |
Collapse
|
11
|
Cui Z, Gong Y, Luo X, Zheng N, Tan S, Liu S, Li Y, Wang Q, Sun F, Hu M, Pan W, Yang X. β-Glucan alleviates goal-directed behavioral deficits in mice infected with Toxoplasma gondii. Parasit Vectors 2023; 16:65. [PMID: 36782332 PMCID: PMC9926625 DOI: 10.1186/s13071-023-05686-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Toxoplasma gondii (T. gondii) is a neuroinvasive parasite causing neuroinflammation, which in turn is associated with a higher risk for several psycho-behavioral disorders. There is an urgent need to identify drugs capable of improving cognitive deficits induced by T. gondii infection. β-Glucan, an active ingredient in mushrooms, could significantly enhance immunity. However, the effects of β-glucan against neuroinflammation and cognitive decline induced by T. gondii infection remain unknown. The present study aimed to investigate the neuroprotective effect of β-glucan on goal-directed behavior of mice chronically infected by T. gondii Wh6 strain. METHODS A mice model of chronic T. gondii Wh6 infection was established by infecting mice by oral gavage with 10 cysts of T. gondii Wh6. Intraperitoneal injection of β-glucan was manipulated 2 weeks before T. gondii infection. Performance of the infected mice on the Y-maze test and temporal order memory (TOM) test was used to assess the goal-directed behavior. Golgi-Cox staining, transmission electron microscopy, immunofluorescence, real-time PCR and western blot assays were used to detect prefrontal cortex-associated pathological change and neuroinflammation. RESULTS The administration of β-glucan significantly prevented T. gondii Wh6-induced goal-directed behavioral impairment as assessed behaviorally by the Y-maze test and TOM test. In the prefrontal cortex, β-glucan was able to counter T. gondii Wh6-induced degeneration of neurites, impairment of synaptic ultrastructure and decrease of pre- and postsynaptic protein levels. Also, β-glucan significantly prevented the hyperactivation of pro-inflammatory microglia and astrocytes, as well as the upregulation of proinflammatory cytokines caused by chronic T. gondii Wh6 infection. CONCLUSIONS This study revealed that β-glucan prevents goal-directed behavioral impairment induced by chronic T. gondii infection in mice. These findings suggest that β-glucan may be an effective drug candidate to prevent T. gondii-associated psycho-behavioral disorders including goal-directed behavioral injury.
Collapse
Affiliation(s)
- Zeyu Cui
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Yuying Gong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Xiaotong Luo
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Niuyi Zheng
- Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Shimin Tan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Shuxi Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Youwei Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Qingling Wang
- Department of Pathology, Basic Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Fenfen Sun
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Minmin Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| |
Collapse
|
12
|
Tao Q, Yang D, Qin K, Liu L, Jin M, Zhang F, Zhu J, Wang J, Luo Q, Du J, Yu L, Shen J, Chu D. Studies on the mechanism of Toxoplasma gondii Chinese 1 genotype Wh6 strain causing mice abnormal cognitive behavior. Parasit Vectors 2023; 16:30. [PMID: 36698166 PMCID: PMC9875435 DOI: 10.1186/s13071-022-05618-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/14/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Alzheimer's disease presents an abnormal cognitive behavior. TgCtwh6 is one of the predominant T. gondii strains prevalent in China. Although T. gondii type II strain infection can cause host cognitive behavioral abnormalities, we do not know whether TgCtwh6 could also cause host cognitive behavioral changes. So, in this study, we will focus on the effect of TgCtwh6 on mouse cognitive behavior and try in vivo and in vitro to explore the underlying mechanism by which TgCtwh6 give rise to mice cognitive behavior changes at the cellular and molecular level. METHODS C57BL/6 mice were infected orally with TgCtwh6 cysts. From day 90 post-infection on, all mice were conducted through the open field test and then Morris water maze test to evaluate cognitive behavior. The morphology and number of cells in hippocampus were examined with hematoxylin-eosin (H&E) and Nissl staining; moreover, Aβ protein in hippocampus was determined with immunohistochemistry and thioflavin S plaque staining. Synaptotagmin 1, apoptosis-related proteins, BACE1 and APP proteins and genes from hippocampus were assessed by western blotting or qRT-PCR. Hippocampal neuronal cell line or mouse microglial cell line was challenged with TgCtwh6 tachyzoites and then separately cultured in a well or co-cultured in a transwell device. The target proteins and genes were analyzed by immunofluorescence staining, western blotting and qRT-PCR. In addition, mouse microglial cell line polarization state and hippocampal neuronal cell line apoptosis were estimated using flow cytometry assay. RESULTS The OFT and MWMT indicated that infected mice had cognitive behavioral impairments. The hippocampal tissue assay showed abnormal neuron morphology and a decreased number in infected mice. Moreover, pro-apoptotic proteins, as well as BACE1, APP and Aβ proteins, increased in the infected mouse hippocampus. The experiments in vitro showed that pro-apoptotic proteins and p-NF-κBp65, NF-κBp65, BACE1, APP and Aβ proteins or genes were significantly increased in the infected HT22. In addition, CD80, pro-inflammatory factors, notch, hes1 proteins and genes were enhanced in the infected BV2. Interestingly, not only the APP and pro-apoptotic proteins in HT22, but also the apoptosis rate of HT22 increased after the infected BV2 were co-cultured with the HT22 in a transwell device. CONCLUSIONS Neuron apoptosis, Aβ deposition and neuroinflammatory response involved with microglia polarization are the molecular and cellular mechanisms by which TgCtwh6 causes mouse cognitive behavioral abnormalities.
Collapse
Affiliation(s)
- Qing Tao
- grid.186775.a0000 0000 9490 772XDepartment of Pathogen Biology, Anhui Province Key Laboratory of Microbiology & Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Di Yang
- grid.186775.a0000 0000 9490 772XDepartment of Pathogen Biology, Anhui Province Key Laboratory of Microbiology & Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Kunpeng Qin
- grid.412679.f0000 0004 1771 3402Department of Orthopaedics, the First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Lei Liu
- grid.59053.3a0000000121679639Department of Blood Transfusion, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Mengmeng Jin
- grid.186775.a0000 0000 9490 772XMaternity and Child Health Hospital of Anhui Province, The Affiliated Maternity and Child Health Hospital of Anhui Medical University, Hefei, China
| | - Famin Zhang
- grid.186775.a0000 0000 9490 772XDepartment of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jinjin Zhu
- grid.186775.a0000 0000 9490 772XDepartment of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jie Wang
- grid.186775.a0000 0000 9490 772XDepartment of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Qingli Luo
- grid.186775.a0000 0000 9490 772XDepartment of Pathogen Biology, Anhui Province Key Laboratory of Microbiology & Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Jian Du
- grid.186775.a0000 0000 9490 772XDepartment of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Li Yu
- grid.186775.a0000 0000 9490 772XDepartment of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jilong Shen
- grid.186775.a0000 0000 9490 772XDepartment of Pathogen Biology, Anhui Province Key Laboratory of Microbiology & Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Deyong Chu
- grid.186775.a0000 0000 9490 772XDepartment of Pathogen Biology, Anhui Province Key Laboratory of Microbiology & Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
Baker TL, Uboldi AD, Tonkin CJ, Wright DK, Vo A, Wilson T, Mychasiuk R, McDonald SJ, Semple BD, Sun M, Shultz SR. Pre-existing Toxoplasma gondii infection increases susceptibility to pentylenetetrazol-induced seizures independent of traumatic brain injury in mice. Front Mol Neurosci 2023; 15:1079097. [PMID: 36683847 PMCID: PMC9849700 DOI: 10.3389/fnmol.2022.1079097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction Post-traumatic epilepsy (PTE) is a debilitating chronic outcome of traumatic brain injury (TBI), and neuroinflammation is implicated in increased seizure susceptibility and epileptogenesis. However, how common clinical factors, such as infection, may modify neuroinflammation and PTE development has been understudied. The neurotropic parasite, Toxoplasma gondii (T. gondii) incurably infects one-third of the world's population. Thus, many TBI patients have a pre-existing T. gondii infection at the time of injury. T. gondii infection results in chronic low-grade inflammation and altered signaling pathways within the brain, and preliminary clinical evidence suggest that it may be a risk factor for epilepsy. Despite this, no studies have considered how a pre-existing T. gondii infection may alter the development of PTE. Methods This study aimed to provide insight into this knowledge gap by assessing how a pre-existing T. gondii infection alters susceptibility to, and severity of, pentylenetetrazol (PTZ)-induced seizures (i.e., a surrogate marker of epileptogenesis/PTE) at a chronic stage of TBI recovery. We hypothesized that T. gondii will increase the likelihood and severity of seizures following PTZ administration, and that this would occur in the presence of intensified neuroinflammation. To test this, 6-week old male and female C57BL/6 Jax mice were intraperitoneally injected with 50,000 T. gondii tachyzoites or with the PBS vehicle only. At 12-weeks old, mice either received a severe TBI via controlled cortical impact or sham injury. At 18-weeks post-injury, mice were administered 40 mg/kg PTZ and video-recorded for evaluation of seizure susceptibility. Fresh cortical tissue was then collected for gene expression analyses. Results Although no synergistic effects were evident between infection and TBI, chronic T. gondii infection alone had robust effects on the PTZ-seizure response and gene expression of markers related to inflammatory, oxidative stress, and glutamatergic pathways. In addition to this, females were more susceptible to PTZ-induced seizures than males. While TBI did not impact PTZ responses, injury effects were evident at the molecular level. Discussion Our data suggests that a pre-existing T. gondii infection is an important modifier of seizure susceptibility independent of brain injury, and considerable attention should be directed toward delineating the mechanisms underlying this pro-epileptogenic factor.
Collapse
Affiliation(s)
- Tamara L. Baker
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Alessandro D. Uboldi
- Division of Infectious Disease and Immune Defense, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Christopher J. Tonkin
- Division of Infectious Disease and Immune Defense, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - David K. Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Anh Vo
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Trevor Wilson
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Stuart J. McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Bridgette D. Semple
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Sandy R. Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia,Health Sciences, Vancouver Island University, Nanaimo, BC, Canada,*Correspondence: Sandy R. Shultz,
| |
Collapse
|
14
|
Effects of diverse Types of Toxoplasma gondii on the outcome of Alzheimer's disease in the rat model. Microb Pathog 2023; 174:105931. [PMID: 36473668 DOI: 10.1016/j.micpath.2022.105931] [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: 09/27/2022] [Revised: 12/02/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Toxoplasma gondii has lifelong persistence in the brain and its cysts can affect gene expression and change diverse biological functions of neurons. Many studies indicated T. gondii infection as a risk factor for the development of behavioral changes and neurodegenerative diseases such as Alzheimer's disease (AD), although the etiopathogenetic link between them has not been exactly elucidated. The current study aimed to examine the effects of chronic toxoplasmosis infection with Types I, II, and III strains (RH, PRU, and VEG) alone and in combination on cognitive impairments and neuronal death in the Aβ1-42-induced rat model of Alzheimer's disease. In the chronic toxoplasmosis phase, Alzheimer's induction was conducted by injecting Aβ1-42 oligomers into the rat brain hippocampus. Behavioral tests were conducted 10 days after the AD induction. Real-time PCR was performed to evaluate T. gondii parasite burden by amplification of the B1 gene. Cytokines IL-1β, TNF-α, and IL-10 were assayed in brain tissue supernatant using ELISA. Also, histopathological examinations were conducted to calculate inflammatory changes and neuronal death in the brain. Our findings showed that chronic toxoplasmosis infection with PRU reduces cognitive disorders, while the RH strain of T. gondii plays a destructive role and aggravates cognitive impairments in AD. Also, infection with a combination of PRU and VEG strains significantly improved spatial learning and memory impairments in Alzheimer's rat model. Histopathological findings also confirmed the results of behavioral tests, so that in AβPRU and AβPRU + VEG groups, neuronal death and infiltration of inflammatory cells were negligible and significantly less than in Alzheimer's and AβRH groups. Our findings indicate that chronic toxoplasmosis infection with PRU strain alone, also in combination with VEG strain can significantly improve cognitive disorders in AD rats, while RH strain plays a destructive role in AD pathogenesis.
Collapse
|
15
|
Rantala MJ, Luoto S, Borráz-León JI, Krams I. Schizophrenia: the new etiological synthesis. Neurosci Biobehav Rev 2022; 142:104894. [PMID: 36181926 DOI: 10.1016/j.neubiorev.2022.104894] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 08/25/2022] [Accepted: 09/25/2022] [Indexed: 10/31/2022]
Abstract
Schizophrenia has been an evolutionary paradox: it has high heritability, but it is associated with decreased reproductive success. The causal genetic variants underlying schizophrenia are thought to be under weak negative selection. To unravel this paradox, many evolutionary explanations have been suggested for schizophrenia. We critically discuss the constellation of evolutionary hypotheses for schizophrenia, highlighting the lack of empirical support for most existing evolutionary hypotheses-with the exception of the relatively well supported evolutionary mismatch hypothesis. It posits that evolutionarily novel features of contemporary environments, such as chronic stress, low-grade systemic inflammation, and gut dysbiosis, increase susceptibility to schizophrenia. Environmental factors such as microbial infections (e.g., Toxoplasma gondii) can better predict the onset of schizophrenia than polygenic risk scores. However, researchers have not been able to explain why only a small minority of infected people develop schizophrenia. The new etiological synthesis of schizophrenia indicates that an interaction between host genotype, microbe infection, and chronic stress causes schizophrenia, with neuroinflammation and gut dysbiosis mediating this etiological pathway. Instead of just alleviating symptoms with drugs, the parasite x genotype x stress model emphasizes that schizophrenia treatment should focus on detecting and treating possible underlying microbial infection(s), neuroinflammation, gut dysbiosis, and chronic stress.
Collapse
Affiliation(s)
- Markus J Rantala
- Department of Biology, University of Turku, FIN-20014 Turku, Finland.
| | - Severi Luoto
- School of Population Health, University of Auckland, 1023 Auckland, New Zealand
| | | | - Indrikis Krams
- Institute of Ecology and Earth Sciences, University of Tartu, 51014 Tartu, Estonia; Department of Zoology and Animal Ecology, Faculty of Biology, University of Latvia, 1004, Rīga, Latvia
| |
Collapse
|
16
|
Ademe M, Kebede T, Teferra S, Alemayehu M, Girma F, Abebe T. Is latent Toxoplasma gondii infection associated with the occurrence of schizophrenia? A case-control study. PLoS One 2022; 17:e0270377. [PMID: 35737701 PMCID: PMC9223392 DOI: 10.1371/journal.pone.0270377] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/08/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction
Neurotropic pathogens such as Toxoplasma gondii (T. gondii) which result in chronic infections in the brain are associated with mental illnesses. In view of this, a growing body of literature has revealed the possible interaction of schizophrenia and T. gondii infection.
Method
A case-control study was conducted from February 2018 to January 2019 among 47 Schizophrenia patients and 47 age and sex-matched controls. Data was collected using a structured questionnaire. Serum was used for serological analysis of anti-T. gondii IgG and IgM antibodies through chemiluminescent immunoassay. Proportions and mean with standard deviations (SD) were used as descriptive measures and variables with p-values <0.05 were considered as statistically significant and independently associated with schizophrenia.
Result
The mean ages of schizophrenia patients and controls were 29.64 ± 5.8 yrs and 30.98 ± 7.3 yrs, respectively. We found that 81.9% (77/94) of the study subjects had a positive anti-T. gondii IgG antibody. While the difference is statistically insignificant, schizophrenic patients have a marginally higher seroprevalence of toxoplasmosis than controls (87.2% vs 80.9%; p = 0.398). Schizophrenia cases who live in homes with soil floors have a significantly higher T. gondii infection as compared to those who live in homes with cement/ceramic floors (90.9% vs 33.3%; p = 0.004). Furthermore, there was a significantly lower T. gondii infection among schizophrenic cases who were taking antipsychotic medication for more than three yrs (79.3% vs 100.0%, p = 0.039). On the other hand, among all study subjects who have T. gondii infection, subjects who are addicted to khat and alcohol were about seven times more likely to develop schizophrenia (71.4% vs 47.7%, OR = 7.13, p = 0.024).
Conclusion
Our data is not sufficient to show a significant positive correlation between T. gondii infection and schizophrenia. For study subjects with T. gondii infection, addiction to khat and alcohol is one of the risk factors for schizophrenia.
Collapse
Affiliation(s)
- Muluneh Ademe
- Department of Microbiology, Immunology and Parasitology, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
- * E-mail:
| | - Tadesse Kebede
- Department of Microbiology, Immunology and Parasitology, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Solomon Teferra
- Department of Psychiatry, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Melkam Alemayehu
- Department of Psychiatry, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Friehiwot Girma
- Department of Pediatrics and Child Health Nursing, School of Health Sciences, College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia
| | - Tamrat Abebe
- Department of Microbiology, Immunology and Parasitology, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
17
|
Calero-Bernal R, Fernández-Escobar M, Katzer F, Su C, Ortega-Mora LM. Unifying Virulence Evaluation in Toxoplasma gondii: A Timely Task. Front Cell Infect Microbiol 2022; 12:868727. [PMID: 35573788 PMCID: PMC9097680 DOI: 10.3389/fcimb.2022.868727] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/28/2022] [Indexed: 01/25/2023] Open
Abstract
Toxoplasma gondii, a major zoonotic pathogen, possess a significant genetic and phenotypic diversity that have been proposed to be responsible for the variation in clinical outcomes, mainly related to reproductive failure and ocular and neurological signs. Different T. gondii haplogroups showed strong phenotypic differences in laboratory mouse infections, which provide a suitable model for mimicking acute and chronic infections. In addition, it has been observed that degrees of virulence might be related to the physiological status of the host and its genetic background. Currently, mortality rate (lethality) in outbred laboratory mice is the most significant phenotypic marker, which has been well defined for the three archetypal clonal types (I, II and III) of T. gondii; nevertheless, such a trait seems to be insufficient to discriminate between different degrees of virulence of field isolates. Many other non-lethal parameters, observed both in in vivo and in vitro experimental models, have been suggested as highly informative, yielding promising discriminatory power. Although intra-genotype variations have been observed in phenotypic characteristics, there is no clear picture of the phenotypes circulating worldwide; therefore, a global overview of T. gondii strain mortality in mice is presented here. Molecular characterization has been normalized to some extent, but this is not the case for the phenotypic characterization and definition of virulence. The present paper proposes a baseline (minimum required information) for the phenotypic characterization of T. gondii virulence and intends to highlight the needs for consistent methods when a panel of T. gondii isolates is evaluated for virulence.
Collapse
Affiliation(s)
- Rafael Calero-Bernal
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
- *Correspondence: Rafael Calero-Bernal, ; Luis Miguel Ortega-Mora,
| | - Mercedes Fernández-Escobar
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| | - Frank Katzer
- Disease Control Department, Moredun Research Institute, Edinburgh, United Kingdom
| | - Chunlei Su
- Department of Microbiology, University of Tennessee, Knoxville, TN, United States
| | - Luis Miguel Ortega-Mora
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
- *Correspondence: Rafael Calero-Bernal, ; Luis Miguel Ortega-Mora,
| |
Collapse
|
18
|
Ponce-Regalado MD, Salazar-Juárez A, Oscar RE, Contis-Montes de Oca A, Hurtado-Alvarado G, Arce-Paredes P, Pérez-Sánchez G, Pavón L, Girón-Pérez MI, Hernández-Pando R, Alvarez-Sánchez ME, Enrique BV. Development of Anxiolytic and Depression-like Behavior in Mice Infected with Mycobacterium lepraemurium. Neuroscience 2022; 493:15-30. [PMID: 35447197 DOI: 10.1016/j.neuroscience.2022.03.021] [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: 05/06/2021] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 11/26/2022]
Abstract
Murine leprosy is a systemic infectious disease of mice caused by Mycobacterium lepraemurium (MLM) in which the central nervous system (CNS) is not infected; nevertheless, diseased animals show measurable cognitive alterations. For this reason, in this study, we explored the neurobehavioral changes in mice chronically infected with MLM. BALB/c mice were infected with MLM, and 120 days later, the alterations in mice were evaluated based on immunologic, histologic, endocrine, neurochemical, and behavioral traits. We found increases in the levels of IL-4 and IL-10 associated with high bacillary loads. We also found increase in the serum levels of corticosterone, epinephrine, and norepinephrine in the adrenal gland, suggesting neuroendocrine deregulation. Mice exhibited depression-like behavior in the tail suspension and forced swimming tests and anxiolytic behavior in the open field and elevated plus maze tests. The neurobehavioral alterations of mice were correlated with the histologic damage in the prefrontal cortex, ventral hippocampus, and amygdala, as well as with a blood-brain barrier disruption in the hippocampus. These results reveal an interrelated response of the neuroimmune-endocrinological axis in unresolved chronic infections that result in neurocognitive deterioration.
Collapse
Affiliation(s)
- M D Ponce-Regalado
- Departamento de Ciencias de la Salud, Centro Universitario de los Altos, Universidad de Guadalajara, Carretera a Yahualica, Km. 7.5 Tepatitlán de Morelos, Jalisco 47600, Mexico
| | - A Salazar-Juárez
- Branch Clinical Research. Laboratory of Molecular Neurobiology and Neurochemistry of Addiction, National Institute of Psychiatry "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - Rojas-Espinosa Oscar
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Carpio y Plan de Ayala, Colonia Santo Tomás, 11340, Ciudad de México, Mexico.
| | - A Contis-Montes de Oca
- Sección de estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Salvador Diaz Miron y Plan de San Luis S/N, Miguel Hidalgo, 11340 Mexico City, Mexico
| | - G Hurtado-Alvarado
- Area of Neurosciences, Department of Biology of Reproduction, Area of Neurosciences, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - P Arce-Paredes
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Carpio y Plan de Ayala, Colonia Santo Tomás, 11340, Ciudad de México, Mexico
| | - G Pérez-Sánchez
- Laboratory of Psychoimmunology, National Institute of Psychiatry "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - L Pavón
- Laboratory of Psychoimmunology, National Institute of Psychiatry "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - M I Girón-Pérez
- Laboratorio de Inmunotoxicología, Secretaría de Investigación y Posgrado Universidad Autónoma de Nayarit, Boulevard Tepic-Xalisco s/n. Cd, de la Cultura Amado Nervo, C.P. 63000 Tepic, Nayarit, México
| | - R Hernández-Pando
- Experimental Pathology Section, Pathology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Vasco de Quiroga 15, Colonia Belisario Dominguez Seccion XVI, 14080, Deleg. Tlalpan, México City, Mexico
| | - M E Alvarez-Sánchez
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), San Lorenzo # 290, Col. Del Valle, CP 03100 México City, México
| | - Becerril-Villanueva Enrique
- Laboratory of Psychoimmunology, National Institute of Psychiatry "Ramón de la Fuente", Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico.
| |
Collapse
|
19
|
Hou Z, Wang L, Su D, Cai W, Zhu Y, Liu D, Huang S, Xu J, Pan Z, Tao J. Global MicroRNAs Expression Profile Analysis Reveals Possible Regulatory Mechanisms of Brain Injury Induced by Toxoplasma gondii Infection. Front Neurosci 2022; 16:827570. [PMID: 35360170 PMCID: PMC8961362 DOI: 10.3389/fnins.2022.827570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/16/2022] [Indexed: 11/13/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is an obligate intracellular parasitic protozoan that can cause toxoplasmosis in humans and other endotherms. T. gondii can manipulate the host gene expression profile by interfering with miRNA expression, which is closely associated with the molecular mechanisms of T. gondii-induced brain injury. However, it is unclear how T. gondii manipulates the gene expression of central nervous system (CNS) cells through modulation of miRNA expression in vivo during acute and chronic infection. Therefore, high-throughput sequencing was used to investigate expression profiles of brain miRNAs at 10, 25, and 50 days post-infection (DPI) in pigs infected with the Chinese I genotype T. gondii strain in this study. Compared with the control group 87, 68, and 135 differentially expressed miRNAs (DEMs) were identified in the infected porcine brains at 10, 25, and 50 DPI, respectively. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis showed that a large number significantly enriched GO terms and KEGG pathways were found, and were mostly associated with stimulus or immune response, signal transduction, cell death or apoptosis, metabolic processes, immune system or diseases, and cancers. miRNA–gene network analysis revealed that the crucial connecting nodes, including DEMs and their target genes, might have key roles in the interactions between porcine brain and T. gondii. These results suggest that the regulatory strategies of T. gondii are involved in the modulation of a variety of host cell signaling pathways and cellular processes, containing unfolded protein response (UPR), oxidative stress (OS), autophagy, apoptosis, tumorigenesis, and inflammatory responses, by interfering with the global miRNA expression profile of CNS cells, allowing parasites to persist in the host CNS cells and contribute to pathological damage of porcine brain. To our knowledge, this is the first report on miRNA expression profile in porcine brains during acute and chronic T. gondii infection in vivo. Our results provide new insights into the mechanisms underlying T. gondii-induced brain injury during different infection stages and novel targets for developing therapeutic agents against T. gondii.
Collapse
Affiliation(s)
- Zhaofeng Hou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Lele Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Dingzeyang Su
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Weimin Cai
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Yu Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Dandan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Siyang Huang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Jinjun Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Zhiming Pan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Jianping Tao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
- *Correspondence: Jianping Tao,
| |
Collapse
|
20
|
Ginsenoside Rh2 reduces depression in offspring of mice with maternal toxoplasma infection during pregnancy by inhibiting microglial activation via the HMGB1/TLR4/NF-κB signaling pathway. J Ginseng Res 2022; 46:62-70. [PMID: 35035240 PMCID: PMC8753429 DOI: 10.1016/j.jgr.2021.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 02/08/2023] Open
Abstract
Background Maternal Toxoplasma gondii (T. gondii) infection during pregnancy has been associated with various mental illnesses in the offspring. Ginsenoside Rh2 (GRh2) is a major bioactive compound obtained from ginseng that has an anti-T. gondii effect and attenuates microglial activation through toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) signaling pathway. GRh2 also alleviated tumor-associated or lipopolysaccharide-induced depression. However, the effects and potential mechanisms of GRh2 on depression-like behavior in mouse offspring caused by maternal T. gondii infection during pregnancy have not been investigated. Methods We examined GRh2 effects on the depression-like behavior in mouse offspring, caused by maternal T. gondii infection during pregnancy, by measuring depression-like behaviors and assaying parameters at the neuronal and molecular level. Results We showed that GRh2 significantly improved behavioral measures: sucrose consumption, forced swim time and tail suspended immobility time of their offspring. These corresponded with increased tissue concentrations of 5-hydroxytryptamine and dopamine, and attenuated indoleamine 2,3-dioxygenase or enhanced tyrosine hydroxylase expression in the prefrontal cortex. GRh2 ameliorated neuronal damage in the prefrontal cortex. Molecular docking results revealed that GRh2 binds strongly to both TLR4 and high mobility group box 1 (HMGB1). Conclusion This study demonstrated that GRh2 ameliorated the depression-like behavior in mouse offspring of maternal T. gondii infection during pregnancy by attenuating the excessive activation of microglia and neuroinflammation through the HMGB1/TLR4/NF-κB signaling pathway. It suggests that GRh2 could be considered a potential therapy in preventing and treating psychiatric disorders in the offspring mice of mothers with prenatal exposure to T. gondii infection.
Collapse
|
21
|
Nayeri T, Sarvi S, Daryani A. Toxoplasmosis: Targeting neurotransmitter systems in psychiatric disorders. Metab Brain Dis 2022; 37:123-146. [PMID: 34476718 DOI: 10.1007/s11011-021-00824-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/14/2021] [Indexed: 12/30/2022]
Abstract
The most common form of the disease caused by Toxoplasma gondii (T. gondii) is latent toxoplasmosis due to the formation of tissue cysts in various organs, such as the brain. Latent toxoplasmosis is probably a risk factor in the development of some neuropsychiatric disorders. Behavioral changes after infection are caused by the host immune response, manipulation by the parasite, central nervous system (CNS) inflammation, as well as changes in hormonal and neuromodulator relationships. The present review focused on the exact mechanisms of T. gondii effect on the alteration of behavior and neurotransmitter levels, their catabolites and metabolites, as well as the interaction between immune responses and this parasite in the etiopathogenesis of psychiatric disorders. The dysfunction of neurotransmitters in the neural transmission is associated with several neuropsychiatric disorders. However, further intensive studies are required to determine the effect of this parasite on altering the level of neurotransmitters and the role of neurotransmitters in the etiology of host behavioral changes.
Collapse
Affiliation(s)
- Tooran Nayeri
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahabeddin Sarvi
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Daryani
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
22
|
Virus MA, Ehrhorn EG, Lui LM, Davis PH. Neurological and Neurobehavioral Disorders Associated with Toxoplasma gondii Infection in Humans. J Parasitol Res 2021; 2021:6634807. [PMID: 34712493 PMCID: PMC8548174 DOI: 10.1155/2021/6634807] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 09/15/2021] [Indexed: 01/17/2023] Open
Abstract
The intracellular parasite Toxoplasma gondii is estimated to infect up to 30% of the world population, leading to lifelong chronic infection of the brain and muscle tissue. Although most latent T. gondii infections in humans have traditionally been considered asymptomatic, studies in rodents suggest phenotypic neurological changes are possible. Consequently, several studies have examined the link between T. gondii infection and diseases such as schizophrenia, epilepsy, depression, bipolar disorder, dysphoria, Alzheimer's disease, Parkinson's disease, and obsessive-compulsive disorder (OCD). To date, there is varying evidence of the relationship of T. gondii to these human neurological or neurobehavioral disorders. A thorough review of T. gondii literature was conducted to highlight and summarize current findings. We found that schizophrenia was most frequently linked to T. gondii infection, while sleep disruption showed no linkage to T. gondii infection, and other conditions having mixed support for a link to T. gondii. However, infection as a cause of human neurobehavioral disease has yet to be firmly established.
Collapse
Affiliation(s)
- Maxwell A. Virus
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Evie G. Ehrhorn
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - LeeAnna M. Lui
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Paul H. Davis
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| |
Collapse
|
23
|
Lan HW, Lu YN, Zhao XD, Jin GN, Lu JM, Jin CH, Ma J, Jin X, Xu X, Piao LX. New role of sertraline against Toxoplasma gondii-induced depression-like behaviours in mice. Parasite Immunol 2021; 43:e12893. [PMID: 34637545 DOI: 10.1111/pim.12893] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 09/27/2021] [Accepted: 10/06/2021] [Indexed: 12/31/2022]
Abstract
Toxoplasma gondii (T. gondii) is a neurotropic protozoan parasite, which can cause mental and behavioural disorders. The present study aimed to elucidate the effects and underlying molecular mechanisms of sertraline (SERT) on T. gondii-induced depression-like behaviours. In the present study, a mouse model and a microglial cell line (BV2 cells) model were established by infecting with the T. gondii RH strain. In in vivo and in vitro experiments, the underlying molecular mechanisms of SERT in inhibiting depression-like behaviours and cellular perturbations caused by T. gondii infection were investigated in the mouse brain and BV2 cells. The administration of SERT significantly ameliorated depression-like behaviours in T. gondii-infected mice. Furthermore, SERT inhibited T. gondii proliferation. Treatment with SERT significantly inhibited the activation of microglia and decreased levels of pro-inflammatory cytokines such as tumour necrosis factor-alpha, and interferon-gamma, by down-regulating tumour necrosis factor receptor 1/nuclear factor-kappa B signalling pathway, thereby ameliorating the depression-like behaviours induced by T. gondii infection. Our study provides insight into the underlying molecular mechanisms of the newly discovered role of SERT against T. gondii-induced depression-like behaviours.
Collapse
Affiliation(s)
- Hui-Wen Lan
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Yu-Nan Lu
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Xu-Dong Zhao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Guang-Nan Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Jing-Mei Lu
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Cheng-Hua Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Juan Ma
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Xuejun Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Xiang Xu
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Lian-Xun Piao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| |
Collapse
|
24
|
Correa Leite PE, de Araujo Portes J, Pereira MR, Russo FB, Martins-Duarte ES, Almeida Dos Santos N, Attias M, Barrantes FJ, Baleeiro Beltrão-Braga PC, de Souza W. Morphological and biochemical repercussions of Toxoplasma gondii infection in a 3D human brain neurospheres model. Brain Behav Immun Health 2021; 11:100190. [PMID: 34589727 PMCID: PMC8474451 DOI: 10.1016/j.bbih.2020.100190] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 12/06/2020] [Indexed: 12/19/2022] Open
Abstract
Background Toxoplasmosis is caused by the parasite Toxoplasma gondii that can infect the central nervous system (CNS), promoting neuroinflammation, neuronal loss, neurotransmitter imbalance and behavioral alterations. T. gondii infection is also related to neuropsychiatric disorders such as schizophrenia. The pathogenicity and inflammatory response in rodents are different to the case of humans, compromising the correlation between the behavioral alterations and physiological modifications observed in the disease. In the present work we used BrainSpheres, a 3D CNS model derived from human pluripotent stem cells (iPSC), to investigate the morphological and biochemical repercussions of T. gondii infection in human neural cells. Methods We evaluated T. gondii ME49 strain proliferation and cyst formation in both 2D cultured human neural cells and BrainSpheres. Aspects of cell morphology, ultrastructure, viability, gene expression of neural phenotype markers, as well as secretion of inflammatory mediators were evaluated for 2 and 4 weeks post infection in BrainSpheres. Results T. gondii can infect BrainSpheres, proliferating and inducing cysts formation, neural cell death, alteration in neural gene expression and triggering the release of several inflammatory mediators. Conclusions BrainSpheres reproduce many aspects of T. gondii infection in human CNS, constituting a useful model to study the neurotoxicity and neuroinflammation mediated by the parasite. In addition, these data could be important for future studies aiming at better understanding possible correlations between psychiatric disorders and human CNS infection with T. gondii. T. gondii infects, proliferates and induce cysts formation in neurospheres. T. gondii infection induces neural cell death in neurospheres. T. gondii infection promotes alteration in neural gene expression in neurospheres. T. gondii infection promotes release of inflammatory mediators in neurospheres.
Collapse
Affiliation(s)
- Paulo Emilio Correa Leite
- Institute of Biophysics Carlos Chagas Filho and National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, RJ, Brazil.,Directory of Metrology Applied to Life Sciences (Dimav), National Institute of Metrology Quality and Technology (INMETRO), Duque de Caxias, RJ, Brazil
| | - Juliana de Araujo Portes
- Institute of Biophysics Carlos Chagas Filho and National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, RJ, Brazil
| | | | - Fabiele Baldino Russo
- Laboratory of Disease Modeling, Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil
| | - Erica S Martins-Duarte
- Institute of Biophysics Carlos Chagas Filho and National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, RJ, Brazil.,Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Nathalia Almeida Dos Santos
- Laboratory of Disease Modeling, Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil.,Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, UK
| | - Marcia Attias
- Institute of Biophysics Carlos Chagas Filho and National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, RJ, Brazil
| | - Francisco J Barrantes
- Laboratory of Molecular Neurobiology, Institute for Biomedical Research (BIOMED), UCA-CONICET, Buenos Aires, Argentina
| | - Patricia Cristina Baleeiro Beltrão-Braga
- Laboratory of Disease Modeling, Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil.,Scientific Platform Pasteur-USP, São Paulo, SP, Brazil
| | - Wanderley de Souza
- Institute of Biophysics Carlos Chagas Filho and National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, RJ, Brazil
| |
Collapse
|
25
|
Mouveaux T, Roger E, Gueye A, Eysert F, Huot L, Grenier-Boley B, Lambert JC, Gissot M. Primary brain cell infection by Toxoplasma gondii reveals the extent and dynamics of parasite differentiation and its impact on neuron biology. Open Biol 2021; 11:210053. [PMID: 34610266 PMCID: PMC8492179 DOI: 10.1098/rsob.210053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Toxoplasma gondii is a eukaryotic parasite that forms latent cysts in the brain of immunocompetent individuals. The latent parasite infection of the immune-privileged central nervous system is linked to most complications. With no drug currently available to eliminate the latent cysts in the brain of infected hosts, the consequences of neurons' long-term infection are unknown. It has long been known that T. gondii specifically differentiates into a latent form (bradyzoite) in neurons, but how the infected neuron responds to the infection remains to be elucidated. We have established a new in vitro model resulting in the production of mature bradyzoite cysts in brain cells. Using dual, host and parasite RNA-seq, we characterized the dynamics of differentiation of the parasite, revealing the involvement of key pathways in this process. Moreover, we identified how the infected brain cells responded to the parasite infection revealing the drastic changes that take place. We showed that neuronal-specific pathways are strongly affected, with synapse signalling being particularly affected, especially glutamatergic synapse signalling. The establishment of this new in vitro model allows investigating both the dynamics of parasite differentiation and the specific response of neurons to long-term infection by this parasite.
Collapse
Affiliation(s)
- Thomas Mouveaux
- U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000 Lille, France
| | - Emmanuel Roger
- U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000 Lille, France
| | - Alioune Gueye
- U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000 Lille, France
| | - Fanny Eysert
- U1167, University of Lille, Inserm, Institut Pasteur de Lille, F-59000 Lille, France
| | - Ludovic Huot
- U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000 Lille, France
| | | | - Jean-Charles Lambert
- U1167, University of Lille, Inserm, Institut Pasteur de Lille, F-59000 Lille, France
| | - Mathieu Gissot
- U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000 Lille, France
| |
Collapse
|
26
|
Pharmacokinetic Properties of the Novel Synthetic Cannabinoid 5F-APINAC and Its Influence on Metabolites Associated with Neurotransmission in Rabbit Plasma. Pharmaceuticals (Basel) 2021; 14:ph14070668. [PMID: 34358094 PMCID: PMC8308683 DOI: 10.3390/ph14070668] [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: 06/09/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 11/23/2022] Open
Abstract
The strong psychoactive effects of synthetic cannabinoids raise the need for the deeper studying of their neurometabolic effects. The pharmacokinetic properties of 5F-APINAC and its influence on metabolomics profiles associated with neurotransmission were investigated in rabbit plasma. Twelve rabbits divided into three groups received 1-mL 5F-APINAC at 0.1, 1 and 2 mg/kg. The intervention groups were compared with the controls. Sampling was performed at nine time points (0–24 h). Ultra-high-performance liquid chromatography–tandem mass spectrometry was used. The pharmacokinetics were dose-dependent (higher curve at a higher dose) with a rapid biotransformation, followed by gradual elimination within 24 h. The tryptophan concentrations abruptly decreased (p < 0.05) in all tested groups, returning to the basal levels after 6 h. 5-hydroxylindole acetic acid increased (p < 0.05) in the controls, but this trend was absent in the treated groups. The aspartic acid concentrations were elevated (p < 0.001) in the treated groups. L-kynurenine was elevated (p < 0.01) in the intervention groups receiving 1 mg/kg to 2 mg/kg. Dose-dependent elevations (p < 0.01) were found for kynurenic acid, xanthurenic acid and quinolinic acid (p < 0.01), whereas the anthranilic acid trends were decreased (p < 0.01). The indole-3-propionic acid and indole-3-carboxaldehyde trends were elevated (p < 0.05), whereas the indole-3-lactic acid trajectories were decreased (p < 0.01) in the intervention groups. 5F-APINAC administration had a rapid biotransformation and gradual elimination. The metabolites related to the kynurenine and serotonergic system/serotonin pathways, aspartic acid innervation system and microbial tryptophan catabolism were altered.
Collapse
|
27
|
Social preference is maintained in mice with impaired startle reflex and glutamate/D-serine imbalance induced by chronic cerebral toxoplasmosis. Sci Rep 2021; 11:14029. [PMID: 34234237 PMCID: PMC8263783 DOI: 10.1038/s41598-021-93504-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/11/2021] [Indexed: 02/06/2023] Open
Abstract
Toxoplasma gondii is an opportunistic protozoan pathogen with a wide geographic distribution. The chronic phase of toxoplasmosis is often asymptomatic in humans and is characterized by tissue cysts throughout the central nervous system and muscle cells. T. gondii and other pathogens with tropism for the central nervous system are considered risk factors in the etiology of several neuropsychiatric disorders, such as schizophrenia and bipolar disorder, besides neurological diseases. Currently, it is known that cerebral toxoplasmosis increases dopamine levels in the brain and it is related to behavioral changes in animals and humans. Here we evaluate whether chronic T. gondii infection, using the cystogenic ME-49 strain, could induce behavioral alterations associated with neuropsychiatric disorders and glutamatergic neurotransmission dysfunction. We observed that the startle amplitude is reduced in the infected animals as well as glutamate and D-serine levels in prefrontal cortical and hippocampal tissue homogenates. Moreover, we did not detect alterations in social preference and spontaneous alternation despite severe motor impairment. Thus, we conclude that behavioral and cognitive aspects are maintained even though severe neural damage is observed by chronic infection of C57Bl/6 mice with the ME-49 strain.
Collapse
|
28
|
Dupont D, Lin JS, Peyron F, Akaoka H, Wallon M. Chronic Toxoplasma gondii infection and sleep-wake alterations in mice. CNS Neurosci Ther 2021; 27:895-907. [PMID: 34085752 PMCID: PMC8265947 DOI: 10.1111/cns.13650] [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: 01/29/2021] [Revised: 03/21/2021] [Accepted: 03/24/2021] [Indexed: 11/29/2022] Open
Abstract
AIM Toxoplasma gondii (Tg) is an intracellular parasite infecting more than a third of the human population. Yet, the impact of Tg infection on sleep, a highly sensitive index of brain functions, remains unknown. We designed an experimental mouse model of chronic Tg infection to assess the effects on sleep-wake states. METHODS Mice were infected using cysts of the type II Prugniaud strain. We performed chronic sleep-wake recordings and monitoring as well as EEG power spectral density analysis in order to assess the quantitative and qualitative changes of sleep-wake states. Pharmacological approach was combined to evaluate the direct impact of the infection and inflammation caused by Tg. RESULTS Infected mouse exhibited chronic sleep-wake alterations over months, characterized by a marked increase (>20%) in time spent awake and in cortical EEG θ power density of all sleep-wake states. Meanwhile, slow-wave sleep decreased significantly. These effects were alleviated by an anti-inflammatory treatment using corticosteroid dexamethasone. CONCLUSION We demonstrated for the first time the direct consequences of Tg infection on sleep-wake states. The persistently increased wakefulness and reduced sleep fit with the parasite's strategy to enhance dissemination through host predation and are of significance in understanding the neurodegenerative and neuropsychiatric disorders reported in infected patients.
Collapse
Affiliation(s)
- Damien Dupont
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.,Physiologie intégrée du système d'éveil, Faculté de Médecine, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Université Claude Bernard Lyon 1, Lyon, France
| | - Jian-Sheng Lin
- Physiologie intégrée du système d'éveil, Faculté de Médecine, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Université Claude Bernard Lyon 1, Lyon, France
| | - François Peyron
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Hideo Akaoka
- Physiologie intégrée du système d'éveil, Faculté de Médecine, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Université Claude Bernard Lyon 1, Lyon, France
| | - Martine Wallon
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.,Physiologie intégrée du système d'éveil, Faculté de Médecine, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
29
|
Ren F, Guo R. Synaptic Microenvironment in Depressive Disorder: Insights from Synaptic Plasticity. Neuropsychiatr Dis Treat 2021; 17:157-165. [PMID: 33519203 PMCID: PMC7838013 DOI: 10.2147/ndt.s268012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022] Open
Abstract
Depression is a major disease that can affect both mental and physical health, limits psychosocial functioning and diminishes the quality of life. But its complex pathogenesis remains poorly understood. The dynamic changes of synaptic structure and function, known as synaptic plasticity, occur with the changes of different cellular microenvironment and are closely related to learning and memory function. Accumulating evidence implies that synaptic plasticity is integrally involved in the pathological changes of mood disorders, especially in depressive disorder. However, the complex dynamic process of synaptic plasticity is influenced by many factors. Here, we reviewed and discussed various factors affecting synaptic plasticity in depression, and proposed a specific framework named synaptic microenvironment, which may be critical for synaptic plasticity under pathological conditions. Based on this concept, we will show how we understand the balance between the synaptic microenvironment and the synaptic plasticity network in depression. Finally, we point out the clinical significance of the synaptic microenvironment in depression.
Collapse
Affiliation(s)
- Feifei Ren
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Rongjuan Guo
- Department of Neurology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, People's Republic of China
| |
Collapse
|
30
|
Meurer YDSR, Brito RMDM, da Silva VP, Andade JMDA, Linhares SSG, Pereira Junior A, de Andrade-Neto VF, de Sá AL, Oliveira CBSD. Toxoplasma gondii infection damages the perineuronal nets in a murine model. Mem Inst Oswaldo Cruz 2020; 115:e200007. [PMID: 32935749 PMCID: PMC7491278 DOI: 10.1590/0074-02760200007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 08/10/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Behavioral and neurochemical alterations associated with toxoplasmosis may be influenced by the persistence of tissue cysts and activation of an immune response in the brain of Toxoplasma gondii-infected hosts. The cerebral extracellular matrix is organised as perineuronal nets (PNNs) that are both released and ensheath by some neurons and glial cells. There is evidences to suggest that PNNs impairment is a pathophysiological mechanism associated with neuropsychiatric conditions. However, there is a lack of information regarding the impact of parasitic infections on the PNNs integrity and how this could affect the host’s behavior. OBJECTIVES In this context, we aimed to analyse the impact of T. gondii infection on cyst burden, PNNs integrity, and possible effects in the locomotor activity of chronically infected mice. METHODS We infected mice with T. gondii ME-49 strain. After thirty days, we assessed locomotor performance of animals using the open field test, followed by evaluation of cysts burden and PNNs integrity in four brain regions (primary and secondary motor cortices, prefrontal and somesthetic cortex) to assess the PNNs integrity using Wisteria floribunda agglutinin (WFA) labeling by immunohistochemical analyses. FINDINGS AND MAIN CONCLUSIONS Our findings revealed a random distribution of cysts in the brain, the disruption of PNNs surrounding neurons in four areas of the cerebral cortex and hyperlocomotor behavior in T. gondii-infected mice. These results can contribute to elucidate the link toxoplasmosis with the establishment of neuroinflammatory response in neuropsychiatric disorders and to raise a discussion about the mechanisms related to changes in brain connectivity, with possible behavioral repercussions during chronic T. gondii infection.
Collapse
Affiliation(s)
- Ywlliane da Silva Rodrigues Meurer
- Universidade Federal da Paraíba, Programa de Pós-Graduação em Neurociência Cognitiva e Comportamento, João Pessoa, PB, Brasil.,Universidade Federal do Rio Grande do Norte, Programa de Pós-Graduação em Psicobiologia, Natal, RN, Brasil
| | - Ramayana Morais de Medeiros Brito
- Universidade Federal do Rio Grande do Norte, Departamento de Microbiologia e Parasitologia, Laboratório de Biologia da Málaria e Toxoplasmose - LABMAT, Natal, RN, Brasil
| | - Valeria Palheta da Silva
- Universidade Federal do Rio Grande do Norte, Programa de Pós-Graduação em Psicobiologia, Natal, RN, Brasil
| | - Joelma Maria de Araujo Andade
- Universidade Federal do Rio Grande do Norte, Departamento de Microbiologia e Parasitologia, Laboratório de Biologia da Málaria e Toxoplasmose - LABMAT, Natal, RN, Brasil
| | | | - Antonio Pereira Junior
- Universidade Federal do Pará, Instituto de Ciências da Sáude, Laboratório de Neuroplasticidade, Belém, PA, Brasil
| | - Valter Ferreira de Andrade-Neto
- Universidade Federal do Rio Grande do Norte, Departamento de Microbiologia e Parasitologia, Laboratório de Biologia da Málaria e Toxoplasmose - LABMAT, Natal, RN, Brasil
| | - Andrea Lima de Sá
- Universidade Federal do Rio Grande do Norte, Departamento de Microbiologia e Parasitologia, Laboratório de Biologia da Málaria e Toxoplasmose - LABMAT, Natal, RN, Brasil
| | - Claudio Bruno Silva de Oliveira
- Universidade Federal do Rio Grande do Norte, Departamento de Microbiologia e Parasitologia, Laboratório de Biologia da Málaria e Toxoplasmose - LABMAT, Natal, RN, Brasil
| |
Collapse
|
31
|
Baker TL, Sun M, Semple BD, Tyebji S, Tonkin CJ, Mychasiuk R, Shultz SR. Catastrophic consequences: can the feline parasite Toxoplasma gondii prompt the purrfect neuroinflammatory storm following traumatic brain injury? J Neuroinflammation 2020; 17:222. [PMID: 32711529 PMCID: PMC7382044 DOI: 10.1186/s12974-020-01885-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/02/2020] [Indexed: 12/02/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality worldwide; however, treatment development is hindered by the heterogenous nature of TBI presentation and pathophysiology. In particular, the degree of neuroinflammation after TBI varies between individuals and may be modified by other factors such as infection. Toxoplasma gondii, a parasite that infects approximately one-third of the world’s population, has a tropism for brain tissue and can persist as a life-long infection. Importantly, there is notable overlap in the pathophysiology between TBI and T. gondii infection, including neuroinflammation. This paper will review current understandings of the clinical problems, pathophysiological mechanisms, and functional outcomes of TBI and T. gondii, before considering the potential synergy between the two conditions. In particular, the discussion will focus on neuroinflammatory processes such as microglial activation, inflammatory cytokines, and peripheral immune cell recruitment that occur during T. gondii infection and after TBI. We will present the notion that these overlapping pathologies in TBI individuals with a chronic T. gondii infection have the strong potential to exacerbate neuroinflammation and related brain damage, leading to amplified functional deficits. The impact of chronic T. gondii infection on TBI should therefore be investigated in both preclinical and clinical studies as the possible interplay could influence treatment strategies.
Collapse
Affiliation(s)
- Tamara L Baker
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Shiraz Tyebji
- Division of Infectious Diseases and Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Christopher J Tonkin
- Division of Infectious Diseases and Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia. .,Department of Medicine, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
32
|
Ghanbari MM, Joneidi M, Kiani B, Babaie J, Sayyah M. Cannabinoid receptors and the proconvulsant effect of toxoplasmosis in mice. Microb Pathog 2020; 144:104204. [DOI: 10.1016/j.micpath.2020.104204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/26/2020] [Accepted: 04/08/2020] [Indexed: 12/29/2022]
|
33
|
Ortiz-Guerrero G, Gonzalez-Reyes RE, de-la-Torre A, Medina-Rincón G, Nava-Mesa MO. Pathophysiological Mechanisms of Cognitive Impairment and Neurodegeneration by Toxoplasma gondii Infection. Brain Sci 2020; 10:brainsci10060369. [PMID: 32545619 PMCID: PMC7349234 DOI: 10.3390/brainsci10060369] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
Toxoplasma gondii is an obligate intracellular parasite considered one of the most successful pathogens in the world, owing to its ability to produce long-lasting infections and to persist in the central nervous system (CNS) in most warm-blooded animals, including humans. This parasite has a preference to invade neurons and affect the functioning of glial cells. This could lead to neurological and behavioral changes associated with cognitive impairment. Although several studies in humans and animal models have reported controversial results about the relationship between toxoplasmosis and the onset of dementia as a causal factor, two recent meta-analyses have shown a relative association with Alzheimer’s disease (AD). AD is characterized by amyloid-β (Aβ) peptide accumulation, neurofibrillary tangles, and neuroinflammation. Different authors have found that toxoplasmosis may affect Aβ production in brain areas linked with memory functioning, and can induce a central immune response and neurotransmitter imbalance, which in turn, affect the nervous system microenvironment. In contrast, other studies have revealed a reduction of Aβ plaques and hyperphosphorylated tau protein formation in animal models, which might cause some protective effects. The aim of this article is to summarize and review the newest data in regard to different pathophysiological mechanisms of cerebral toxoplasmosis and their relationship with the development of AD and cognitive impairment. All these associations should be investigated further through clinical and experimental studies.
Collapse
Affiliation(s)
- Gloria Ortiz-Guerrero
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Rodrigo E. Gonzalez-Reyes
- GI en Neurociencias-NeURos, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia; (R.E.G.-R.); (A.d.-l.-T.); (G.M.-R.)
| | - Alejandra de-la-Torre
- GI en Neurociencias-NeURos, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia; (R.E.G.-R.); (A.d.-l.-T.); (G.M.-R.)
| | - German Medina-Rincón
- GI en Neurociencias-NeURos, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia; (R.E.G.-R.); (A.d.-l.-T.); (G.M.-R.)
| | - Mauricio O. Nava-Mesa
- GI en Neurociencias-NeURos, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia; (R.E.G.-R.); (A.d.-l.-T.); (G.M.-R.)
- Correspondence: ; Tel.: +57-1-2970200 (ext. 3354); Fax: +571-3440351
| |
Collapse
|
34
|
El Mouhawass A, Hammoud A, Zoghbi M, Hallit S, Haddad C, El Haddad K, El Khoury S, Tannous J, Obeid S, Halabi MA, Mammari N. Relationship between Toxoplasma gondii seropositivity and schizophrenia in the Lebanese population: potential implication of genetic polymorphism of MMP-9. BMC Psychiatry 2020; 20:264. [PMID: 32460746 PMCID: PMC7254747 DOI: 10.1186/s12888-020-02683-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/18/2020] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Toxoplasma multiplication and its persistence into the brain cause a local neuroinflammatory reaction, resulting synthesis of neurotransmitters involved in neurological disorders, especially schizophrenia. The Matrix metallopeptidase 9 (MMP-9) protein can play a major role in this neuroinflammation. It can promote extravasation and migration of infected immune cells into the brain. The objectives of this study are to determine the possible association between schizophrenia and toxoplasmosis and highlight the existence of gene polymorphism encoding MMP-9 protein's in patients presented both schizophrenia and toxoplasmosis. METHODS A case-control study was conducted on 150 patients with schizophrenia (case group), and 150 healthy persons (control group). Groups were matched with age, gender, and place of residence. The survey was conducted using a questionnaire and a serological profile assay for specific IgG and IgM antibodies against T. gondii. Reverse transcription-polymerase chain reaction (RT-PCR) of gene polymorphism encoding MMP-9 was performed on 83 cases selected randomly. RESULTS Data show a significant association between toxoplasmosis (IgM+/IgG+ serological profile) and schizophrenia. Significant effects of raw meat consumption and contact with cats have been associated with the occurrence of schizophrenia. RT-PCR shows the presence of muted allele of MMP-9 gene in selected cases whose present T. gondii serological profile IgM+/IgG+ and IgM-/IgG+ respectively. CONCLUSION Toxoplasmosis may be one of the etiological causes of schizophrenia, and MMP-9 gene polymorphism could be involved in the occurrence mechanism of this pathology following Toxoplasma infection.
Collapse
Affiliation(s)
- Amata El Mouhawass
- Medical Laboratory Department, Holy Family University, Batroun, 5534 Lebanon
| | - Amale Hammoud
- Public Health Faculty, Jinan University, Tripoli, Lebanon
| | - Marouan Zoghbi
- Psychiatric Hospital of the Cross, Jal Eddib, 6096 Lebanon
- Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Souheil Hallit
- Faculty of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Jounieh, Lebanon
- INSPECT-LB: Institut National de Santé Publique, Épidémiologie Clinique et Toxicologie, Beirut, Lebanon
| | - Chadia Haddad
- Psychiatric Hospital of the Cross, Jal Eddib, 6096 Lebanon
- INSERM, Univ. Limoges, CH Esquirol Limoges, IRD, U1094 Tropical Neuroepidemiology, Institute of Epidemiology and Tropical Neurology, GEIST, Limoges, France
| | - Kinda El Haddad
- Medical Laboratory Department, Holy Family University, Batroun, 5534 Lebanon
| | - Saydeh El Khoury
- Medical Laboratory Department, Holy Family University, Batroun, 5534 Lebanon
| | - Jennifer Tannous
- Medical Laboratory Department, Holy Family University, Batroun, 5534 Lebanon
| | - Sahar Obeid
- Psychiatric Hospital of the Cross, Jal Eddib, 6096 Lebanon
- INSPECT-LB: Institut National de Santé Publique, Épidémiologie Clinique et Toxicologie, Beirut, Lebanon
- Faculty of Arts and Sciences, Holy Spirit University of Kaslik (USEK), Jounieh, Lebanon
| | | | - Nour Mammari
- Medical Laboratory Department, Holy Family University, Batroun, 5534 Lebanon
| |
Collapse
|
35
|
Sun X, Wang T, Wang Y, Ai K, Pan G, Li Y, Zhou C, He S, Cong H. Downregulation of lncRNA-11496 in the Brain Contributes to Microglia Apoptosis via Regulation of Mef2c in Chronic T. gondii Infection Mice. Front Mol Neurosci 2020; 13:77. [PMID: 32499679 PMCID: PMC7243434 DOI: 10.3389/fnmol.2020.00077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/20/2020] [Indexed: 01/02/2023] Open
Abstract
Though it is well known that chronic infections of Toxoplasma gondii (T. gondii) can induce mental and behavioral disorders in the host, little is known about the role of long non-coding RNAs (lncRNAs) in this pathological process. In this study, we employed an advanced lncRNAs and mRNAs integration chip (Affymetrix HTA 2.0) to detect the expression of both lncRNAs and mRNAs in T. gondii Chinese 1 strain infected mouse brain. As a result, for the first time, the downregulation of lncRNA-11496 (NONMMUGO11496) was identified as the responsible factor for this pathological process. We showed that dysregulation of lncRNA-11496 affected proliferation, differentiation and apoptosis of mouse microglia. Furthermore, we proved that Mef2c (Myocyte-specific enhancer factor 2C), a member of the MEF2 subfamily, is the target gene of lncRNA-11496. In a more detailed study, we confirmed that lncRNA-11496 positively regulated the expression of Mef2c by binding to histone deacetylase 2 (HDAC2). Importantly, Mef2c itself could coordinate neuronal differentiation, survival, as well as synapse formation. Thus, our current study provides the first evidence in terms of the modulatory action of lncRNAs in chronic toxoplasmosis in T. gondii infected mouse brain, providing a solid scientific basis for using lncRNA-11496 as a therapeutic target to treat T. gondii induced neurological disorder.
Collapse
Affiliation(s)
- Xiahui Sun
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ting Wang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yongliang Wang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Kang Ai
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ge Pan
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yan Li
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunxue Zhou
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shenyi He
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hua Cong
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
36
|
de Medeiros Brito RM, da Silva Rodrigues Meurer Y, da Silva Santos L, de Melo Marcelino BM, de Andrade-Neto VF. Chronic Toxoplasma gondii infection contributes to decreasing of perineuronal nets surrounding neurons in the Corpus striatum of mice. Parasitol Res 2020; 119:1989-1995. [PMID: 32291469 DOI: 10.1007/s00436-020-06674-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/20/2020] [Indexed: 01/06/2023]
Abstract
Recent advances in chronic toxoplasmosis understanding became the focus of discussion about behavioral abnormalities, which could be explained by cyst location and neuronal impairment in specific brain areas. Perineuronal nets (PNNs) are specialized extracellular matrices that surround the neuronal body and proximal dendrites and play key roles in neuronal circuitry maintenance and stabilization. Its impairment can lead to abnormal synaptic functioning with behavioral repercussions. In this context, we analyzed the impact of Toxoplasma gondii infection on neuronal integrity in the Corpus striatum of chronically infected mice. C57BL/6 and Balb/c female mice were infected with T. gondii ME49 cysts. Brain sections were submitted to immunohistochemistry with Wisteria floribunda agglutinin (WFA) for PNN labeling followed by quantification of tissue cyst and labeled neuronal cells 30 days after infection. Our results revealed that C57BL/6 exhibited a significant decrease in PNN-positive (WFA+) labeled neurons and an expressively higher number of tissue cysts than Balb/c mice. It was also possible to observe that the number of T. gondii tissue cysts and the number of WFA+ neurons were inversely correlated for C57BL/6-infected mice. However, no correlation was observed for Balb/c mice. These data suggest how the impact of parasite dissemination in the brain and host characteristics can influence neuronal integrity impairment during infection by decreasing WFA+ neurons. This might be a plausible pathway in which the presence of T. gondii contributes to behavioral changes in the infected host.
Collapse
Affiliation(s)
- Ramayana Morais de Medeiros Brito
- Graduate Program in Parasitary Biology, Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil.,Laboratory of Malaria and Toxoplasmosis Biology, Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Ywlliane da Silva Rodrigues Meurer
- Laboratory of Malaria and Toxoplasmosis Biology, Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil.,Laboratory of Behavioral Neuroscience, Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil
| | - Lidiane da Silva Santos
- Laboratory of Malaria and Toxoplasmosis Biology, Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Brenna Marceliane de Melo Marcelino
- Laboratory of Malaria and Toxoplasmosis Biology, Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil.,Graduate Program in Biological Sciences, Biosciences Centre, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Valter Ferreira de Andrade-Neto
- Graduate Program in Parasitary Biology, Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil. .,Laboratory of Malaria and Toxoplasmosis Biology, Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil. .,Graduate Program in Biological Sciences, Biosciences Centre, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil.
| |
Collapse
|
37
|
Abstract
Toxoplasma gondii is a ubiquitous, intracellular protozoan parasite with a broad range of intermediate hosts, including humans and rodents. In many hosts, T. gondii establishes a latent long-term infection by converting from its rapidly dividing or lytic form to its slowly replicating and encysting form. In humans and rodents, the major organ for encystment is the central nervous system (CNS), which has led many to investigate how this persistent CNS infection might influence rodent and human behavior and, more recently, neurodegenerative diseases. Toxoplasma gondii is a ubiquitous, intracellular protozoan parasite with a broad range of intermediate hosts, including humans and rodents. In many hosts, T. gondii establishes a latent long-term infection by converting from its rapidly dividing or lytic form to its slowly replicating and encysting form. In humans and rodents, the major organ for encystment is the central nervous system (CNS), which has led many to investigate how this persistent CNS infection might influence rodent and human behavior and, more recently, neurodegenerative diseases. Given the interest in this topic, here we seek to take a global approach to the data for and against the effects of latent T. gondii on behavior and neurodegeneration and the proposed mechanisms that might underlie behavior modifications.
Collapse
|
38
|
Cheng JH, Xu X, Li YB, Zhao XD, Aosai F, Shi SY, Jin CH, Piao JS, Ma J, Piao HN, Jin XJ, Piao LX. Arctigenin ameliorates depression-like behaviors in Toxoplasma gondii-infected intermediate hosts via the TLR4/NF-κB and TNFR1/NF-κB signaling pathways. Int Immunopharmacol 2020; 82:106302. [PMID: 32086097 DOI: 10.1016/j.intimp.2020.106302] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/27/2020] [Accepted: 02/10/2020] [Indexed: 01/23/2023]
Abstract
Toxoplasma gondii (T. gondii) is a known neurotropic protozoan that remains in the central nervous system and induces neuropsychiatric diseases in intermediate hosts. Arctigenin (AG) is one of the major bioactive lignans of the fruit Arctium lappa L. and has a broad spectrum of pharmacological activities such as neuroprotective, anti-inflammatory and anti-T. gondii effects. However, the effect of AG against depressive behaviors observed in T. gondii-infected hosts has not yet been clarified. In the present study, we analyzed the effects of AG against T. gondii-induced depressive behaviors in intermediate hosts using a microglia cell line (BV2 cells) and brain tissues of BALB/c mice during the acute phase of infection with the RH strain of T. gondii. AG attenuated microglial activation and neuroinflammation via the Toll-like receptor/nuclear factor-kappa B (NF-κB) and tumor necrosis factor receptor 1/NF-κB signaling pathways, followed by up-regulating the dopamine and 5-hydroxytryptamine levels and inhibiting the depression-like behaviors of hosts. AG also significantly decreased the T. gondii burden in mouse brain tissues. In conclusion, we elucidated the effects and underlying molecular mechanisms of AG against depressive behaviors induced by T. gondii infection.
Collapse
Affiliation(s)
- Jia-Hui Cheng
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Xiang Xu
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Ying-Biao Li
- Department of Neurology, Affliated Hospital of Yanbian University, Yanji 133000, Jilin, China
| | - Xu-Dong Zhao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Fumie Aosai
- Department of Infection and Host Defense, Graduate School of Medicine, Shinshu University, Matsumoto, Japan
| | - Su-Yun Shi
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Cheng-Hua Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Jing-Shu Piao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Juan Ma
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Hu-Nan Piao
- Department of Neurology, Affliated Hospital of Yanbian University, Yanji 133000, Jilin, China.
| | - Xue-Jun Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China.
| | - Lian-Xun Piao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China. https://orcid.org/0000-0002-8315-5918
| |
Collapse
|
39
|
Boillat M, Hammoudi PM, Dogga SK, Pagès S, Goubran M, Rodriguez I, Soldati-Favre D. Neuroinflammation-Associated Aspecific Manipulation of Mouse Predator Fear by Toxoplasma gondii. Cell Rep 2020; 30:320-334.e6. [PMID: 31940479 PMCID: PMC6963786 DOI: 10.1016/j.celrep.2019.12.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/27/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
Abstract
In rodents, the decrease of felid aversion induced by Toxoplasma gondii, a phenomenon termed fatal attraction, is interpreted as an adaptive manipulation by the neurotropic protozoan parasite. With the aim of understanding how the parasite induces such specific behavioral modifications, we performed a multiparametric analysis of T. gondii-induced changes on host behavior, physiology, and brain transcriptome as well as parasite cyst load and distribution. Using a set of complementary behavioral tests, we provide strong evidence that T. gondii lowers general anxiety in infected mice, increases explorative behaviors, and surprisingly alters predator aversion without selectivity toward felids. Furthermore, we show a positive correlation between the severity of the behavioral alterations and the cyst load, which indirectly reflects the level of inflammation during brain colonization. Taken together, these findings refute the myth of a selective loss of cat fear in T. gondii-infected mice and point toward widespread immune-related alterations of behaviors.
Collapse
Affiliation(s)
- Madlaina Boillat
- Department of Genetics and Evolution, Faculty of Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Pierre-Mehdi Hammoudi
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva CMU, 1 rue Michel-Servet 1211 Geneva 4, Switzerland
| | - Sunil Kumar Dogga
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva CMU, 1 rue Michel-Servet 1211 Geneva 4, Switzerland
| | - Stéphane Pagès
- Wyss Center for Bio- and Neuroengineering, Geneva, Switzerland; Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Maged Goubran
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Ivan Rodriguez
- Department of Genetics and Evolution, Faculty of Sciences, University of Geneva, 1211 Geneva, Switzerland.
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva CMU, 1 rue Michel-Servet 1211 Geneva 4, Switzerland.
| |
Collapse
|
40
|
Guanabenz Reverses a Key Behavioral Change Caused by Latent Toxoplasmosis in Mice by Reducing Neuroinflammation. mBio 2019; 10:mBio.00381-19. [PMID: 31040237 PMCID: PMC6495372 DOI: 10.1128/mbio.00381-19] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Toxoplasma gondii is an intracellular parasite that has infected one-third of humans. The infection is permanent because the replicative form (tachyzoite) converts into a latent tissue cyst form (bradyzoite) that evades host immunity and is impervious to current drugs. The continued presence of these parasitic cysts hinders treatment and leads to chronic infection that has been linked to behavioral changes in rodents and neurological disease in humans. How these behavioral changes occur, and whether they are due to parasite manipulation or the host response to infection, remains an outstanding question. We previously showed that guanabenz possesses antiparasitic activity; here, we show that guanabenz reproducibly lowers brain cyst burden up to 80% in chronically infected male and female BALB/cJ mice when given intraperitoneally but not when administered by gavage or in food. Regardless of the administration route, guanabenz reverses Toxoplasma-induced hyperactivity in latently infected mice. In contrast, guanabenz increases cyst burden when given to chronically infected C57BL/6J mice yet still reverses Toxoplasma-induced hyperactivity. Examination of the brains from chronically infected BALB/cJ and C57BL/6J mice shows that guanabenz decreases inflammation and perivascular cuffing in each strain. Our study establishes a robust model for cyst reduction in BALB/cJ mice and shows for the first time that it is possible to reverse a key behavioral change associated with latent toxoplasmosis. The rescue from parasite-induced hyperactivity correlates with a decrease in neuroinflammation rather than reduced cyst counts, suggesting that some behavioral changes arise from host responses to infection.IMPORTANCE Toxoplasma gondii is a common parasite of animals, including up to one-third of humans. The single-celled parasite persists within hosts for the duration of their life as tissue cysts, giving rise to chronic infection. Latent toxoplasmosis is correlated with neurological dysfunction in humans and results in dramatic behavioral changes in rodents. When infected, mice and rats adapt behaviors that make them more likely to be devoured by cats, the only host that supports the sexual stage of the parasite. In this study, we establish a new mouse model of tissue cyst depletion using a drug called guanabenz and show that it is possible to reverse a key behavior change back to normal in infected animals. We also show that the mechanism appears to have nothing to do with parasite cyst burden but rather the degree of neuroinflammation produced by chronic infection.
Collapse
|
41
|
Vondroušová J, Mikoška M, Syslová K, Böhmová A, Tejkalová H, Vacek L, Kodym P, Krsek D, Horáček J. Monitoring of kynurenine pathway metabolites, neurotransmitters and their metabolites in blood plasma and brain tissue of individuals with latent toxoplasmosis. J Pharm Biomed Anal 2019; 170:139-152. [PMID: 30925271 DOI: 10.1016/j.jpba.2019.03.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/14/2019] [Accepted: 03/17/2019] [Indexed: 12/17/2022]
Abstract
The aim of the presented work was to develop a highly sensitive, accurate and rapid analytical method for the determination of concentration levels of tryptophan and its metabolites of kynurenine catabolic pathway, as well as neurotransmitters and their metabolites in complex biological matrices (brain tissue and blood plasma). The developed analytical method consists of analytes separation from the biological matrices by protein precipitation (blood plasma) or solvent extraction (brain tissue), derivatization of the analytes and their detection by high-performance liquid chromatography combined with mass spectrometry. Individual steps of the whole process were optimized and the method was validated in the terms of selectivity, linearity (R2≥0.980), precision (RSD ≤ 13.3%), recovery (≥82.0%), limit of detection (1.8 ng/mL of blood plasma, 2.2 pg/mg of brain tissue) and limit of quantification (2.5 ng/mL of blood plasma, 2.8 pg/mg of brain tissue). The method was subsequently verified by an animal study, where the concentration levels of the analytes in biological matrices (blood plasma and brain tissue) of T. gondii - infected rats and control animals were compared. All the data obtained from the animal study were statistically evaluated. Increased concentration levels of kynurenine catabolic pathway metabolites (e.g. kynurenine, 3-hydroxykynurenine, quinolinic acid) were observed in the case of T. gondii - infected rats in contrast to the control group. The opposite effect was determined in the case of serotonin and its metabolite 5-hydroxyindoleacetic acid, where higher concentration levels were found in blood plasma of healthy subjects. Finally, Principal Component Analysis (PCA) was utilized for a score plot formation. PCA score plots have demonstrated the similarities of individuals within each group and the differences among the groups.
Collapse
Affiliation(s)
- Jana Vondroušová
- Department of Organic Technology, University of Chemistry and Technology Prague, Technická 5, 166 28, Prague 6, Czech Republic
| | - Miloš Mikoška
- Department of Organic Technology, University of Chemistry and Technology Prague, Technická 5, 166 28, Prague 6, Czech Republic
| | - Kamila Syslová
- Department of Organic Technology, University of Chemistry and Technology Prague, Technická 5, 166 28, Prague 6, Czech Republic.
| | - Adéla Böhmová
- Department of Organic Technology, University of Chemistry and Technology Prague, Technická 5, 166 28, Prague 6, Czech Republic
| | - Hana Tejkalová
- National Institute of Mental Health, Topolová 748, 250 67, Klecany, Czech Republic
| | - Lukáš Vacek
- Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic
| | - Petr Kodym
- The National Institute of Public Health, Šrobárova 48, 100 42, Prague 10, Czech Republic
| | - Daniel Krsek
- The National Institute of Public Health, Šrobárova 48, 100 42, Prague 10, Czech Republic
| | - Jiří Horáček
- National Institute of Mental Health, Topolová 748, 250 67, Klecany, Czech Republic
| |
Collapse
|
42
|
The Association between Toxoplasma gondii Infection and Risk of Parkinson's Disease: A Systematic Review and Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8186017. [PMID: 30931331 PMCID: PMC6413381 DOI: 10.1155/2019/8186017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/20/2019] [Accepted: 02/12/2019] [Indexed: 02/08/2023]
Abstract
Background Several studies have investigated the association between Toxoplasma gondii (T. gondii) infection and risk of Parkinson's disease (PD) with inconsistent results. Clarifying this relation might be useful for better understanding of the risk factors and the relevant mechanisms of PD, thus a meta-analysis was conducted to explore whether exposure to T. gondii is associated with an increased risk of PD. Methods We conducted this meta-analysis according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. A rigorous literature selection was performed by using the databases of PubMed, Embase, Web of Science, Cochrane Library, and ScienceDirect. Odds ratio (OR) and corresponding 95% confidential interval (CI) were pooled by using fixed-effects models. Sensitivity analysis, publication bias test, and methodological quality assessment of studies were also performed. Results Seven studies involving 1086 subjects were included in this meta-analysis. Pooled data by using fixed-effects models suggested both latent infection (OR, 1.17; 95% CI, 0.86 to 1.58; P=0.314) and acute infection (OR, 1.13; 95% CI, 0.30 to 4.35; P=0.855) were not associated with PD risk. Stable and robust estimates were confirmed by sensitivity analysis. No publication bias was found by visual inspection of the funnel plot, Begg's, and Egger's test. Conclusions This meta-analysis does not support any possible association between T. gondii infection and risk of PD. Researches are still warranted to further explore the underlying mechanisms of T. gondii in the pathogenesis of PD and their causal relationship.
Collapse
|